WO2019092939A1 - Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease - Google Patents

Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease Download PDF

Info

Publication number
WO2019092939A1
WO2019092939A1 PCT/JP2018/028998 JP2018028998W WO2019092939A1 WO 2019092939 A1 WO2019092939 A1 WO 2019092939A1 JP 2018028998 W JP2018028998 W JP 2018028998W WO 2019092939 A1 WO2019092939 A1 WO 2019092939A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
spinal cord
cell
serum
cord injury
Prior art date
Application number
PCT/JP2018/028998
Other languages
French (fr)
Japanese (ja)
Inventor
博道 仁科
博子 矢永
Original Assignee
株式会社リジェネシスサイエンス
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 株式会社リジェネシスサイエンス filed Critical 株式会社リジェネシスサイエンス
Priority to JP2019551884A priority Critical patent/JP7207742B2/en
Priority to US16/763,023 priority patent/US20210363483A1/en
Priority to EP18876286.8A priority patent/EP3708662A4/en
Priority to AU2018364852A priority patent/AU2018364852B2/en
Priority to RU2019110931A priority patent/RU2730864C1/en
Priority to CN201880082876.1A priority patent/CN111566204B/en
Priority to CA3080691A priority patent/CA3080691C/en
Publication of WO2019092939A1 publication Critical patent/WO2019092939A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/36Skin; Hair; Nails; Sebaceous glands; Cerumen; Epidermis; Epithelial cells; Keratinocytes; Langerhans cells; Ectodermal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • A61K38/063Glutathione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/185Nerve growth factor [NGF]; Brain derived neurotrophic factor [BDNF]; Ciliary neurotrophic factor [CNTF]; Glial derived neurotrophic factor [GDNF]; Neurotrophins, e.g. NT-3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/01Preparation of mutants without inserting foreign genetic material therein; Screening processes therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0018Culture media for cell or tissue culture
    • C12N5/0031Serum-free culture media
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0622Glial cells, e.g. astrocytes, oligodendrocytes; Schwann cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0625Epidermal cells, skin cells; Cells of the oral mucosa
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/13Nerve growth factor [NGF]; Brain-derived neurotrophic factor [BDNF]; Cilliary neurotrophic factor [CNTF]; Glial-derived neurotrophic factor [GDNF]; Neurotrophins [NT]; Neuregulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/999Small molecules not provided for elsewhere

Definitions

  • the present invention relates to a method for producing cells obtained by culturing cells obtained from human skin tissue for treating a spinal cord injury disease group, and cells obtained by the above-mentioned method or a secretion such as culture medium or Exosome.
  • the present invention relates to a method for producing a medical drug for treating a spinal cord injury disease group containing
  • Spinal cord injury is a condition in which the spine is damaged mainly by applying a strong external force to the spinal column, and the spinal cord is damaged. Similar disorders also occur with internal causes such as spinal cord tumors, hernias and cervical spinal myelopathy. The estimated number of patients with spinal cord injury is 100,000 in Japan. Most of the causes of spinal cord injuries are injuries due to traffic or falling from high places.
  • the central nervous system including the spinal cord can not be repaired or regenerated once it is damaged. Regeneration of the destroyed spinal cord and regaining its function are the wishes of spinal cord all over the world, and research is underway in various fields.
  • a large amount of steroid agent administration is performed within 48 to 72 hours immediately after injury.
  • their bone marrow fluid is cultured and injected into the spinal cord, and groups such as Keio University in Japan have injured the spinal cord within 78 hours after an accident, etc.
  • Attempts have been made to administer hepatocyte growth factor (HFG) and the like.
  • HFG hepatocyte growth factor
  • rehabilitation is performed.
  • rehabilitation of spinal cord injury is not to restore lost function. That is because it is impossible if the nerve does not regenerate.
  • the purpose of rehabilitation is "how to use the remaining functions and enable ADL (Daily life movement)".
  • complications such as hemorrhoids and urinary tract infections occur in the chronic stage, and care becomes extremely difficult.
  • Non-Patent Document 13 human skin-derived stem cells (Mesenchymal stem cells) are difficult to induce in neurons after transplantation and are likely to become chondrocytes and osteocytes, and brain-derived neural stem cells have disadvantages such as low therapeutic effect (Non-Patent Document 13).
  • Non-patent Document 14 It has been reported that precursor cells from the skin are more effective in treating spinal cord injury than neural stem cells from the brain.
  • GDNF glial cell line-derived neurotrophic factor
  • Non-patent Document 8-10 an attempt to further create a healing effect of spinal cord injury is an attempt to further introduce the gene of GDNF into cells expected to have a healing effect.
  • Olfactory ensheathing cells, Schwann cells, etc. are used, and since these cells themselves have a healing effect, there is a further increase in the therapeutic effect.
  • the drawback of this method is that the gene is transferred using a virus, so there is a high possibility of becoming cancerous when transplanted into a living body.
  • Non-patent Document 15 GDNF protein itself is unstable and difficult to handle.
  • Non-patent Document 16 GDNF has also been reported as an intrinsic factor required for the recovery of spinal cord injury.
  • Non-patent Literature 17-18 GDNF has also been reported to be effective in suppressing allodynia that afflicts patients with spinal cord injury.
  • Non-patent Document 19 GDNF has also been reported to be effective in chronic spinal cord injury.
  • Non-patent Document 20 expressing GDNF in cells to be transplanted leads to a breakthrough in the therapeutic effect of spinal cord injury.
  • Tsuji O, et al . Therapeutic potential of appropriately evaluated self-induced pluripotent stem cells for spinal cord injury.
  • PNAS 2010; 107 (28), 12704-12709 Inoue H, et al .: Improvement of hind-paralysis following traumatic spinal cord injury in rats by grafting normal human keratinocytes: new cell-therapy strategy for nerve regeneration.
  • Inoue H, et al . Improvement of hind-limb paralysis followed traumatic spinal cord injury in rats by grafting normal human keratinocytes: new cell-therapy strategy for nerve regeneration.
  • J Artif Organs. J Artif Organs.
  • Deng LX, et al . A novel growth-promoting pathway formed by GDNF-overexpressing Schwann cells promotes propriospinal axonal regeneration, synapse formation, and partial recovery of function after spinal cord injury. 5655-67. Stephen J. A. Daviees, et al .: Transplantaion of specific human astrocytes promotes functional recovery after spinal cord injury. PLoS ONE 2011; 6 (3): e17328 Sowa NA, et al .: Ecto-5'-nucleotidase (CD 73) inhibitors nociception by hydrolyzing AMP to adenosine in nociceptive circuits. J Neurosci.
  • MR et al .
  • Acute exercise prevent the development of neuropathic pain and the sprouting of non-peptidergic (GDNF- and artemin-responsive) c-fibers after spinal cord injury.
  • GDNF non-peptidergic
  • Dolbeare D et al .: Restriction of axonal retraction and promotion of axonal regeneration by chronically injured neurons after intraspinal treatment with glial cell line-derived neurotrophic factor (GDNF). J Neurotrauma. 2003; 20 (11): 1251-61.
  • Lu Y, et al . Glial cell line-derived neurotrophic factor-transfected placenta-derived versus bone marrow-derived mesenchymal cells for treating spinal cord injury. Med Sci Monit. 2017; 23: 1800-1811 Codega P, et al .: Prospective identification and purification of quiescent adult neural stem cells from their in vivo niche. Neuron. 2014; 82 (3): 545-59. DeCarolis NA, et al .: In vivo contribution of nestin-and GLAST-lineage cells to adult hippocampal neurogenesis. 2013; 23 (8): 708-719. Sufan Wu, et al .: Bone marrow stromal cells enhancement differentiation of cocultured neurosphere cells and promote regeneration of injured spinal cord. J Neurosci Res. 2003; 72 (3): 343-51.
  • An object of the present invention is to provide a cell culture method for safely increasing GDNF gene expression (mRNA) without adopting viral gene transfer method.
  • An object of the present invention is to provide a cell culture method which also expresses various markers such as markers of neural stem cells.
  • An object of the present invention is to provide a method for producing a therapeutic agent for spinal cord injury.
  • the present invention is a serum-free medium containing human skin-derived stem cells and any one or more of SAG, palmorphamine and sonic hedgehog (SHH) proteins. It is based on the findings from the example that by culturing, glial cell line-derived neurotrophic factor (GDNF) mRNA can be highly expressed (for example, 10 times or more of that in normal culture).
  • GDNF glial cell line-derived neurotrophic factor
  • a first aspect of the present invention relates to a method for producing a cultured cell containing glial cell line-derived neurotrophic factor (GDNF) mRNA.
  • This method involves culturing a human skin-derived stem cell in a serum-free medium containing one or more of SAG, palmorphamine (Purmorphamine) and Sonic hedgehog (SHH) proteins. Including.
  • SAG palmorphamine
  • SHH Sonic hedgehog
  • This method highly expresses glial cell line-derived neurotrophic factor (GDNF) mRNA.
  • Human skin-derived stem cells are preferably derived from skin cells of the patient himself. In particular, it is preferable to collect cells from skin tissue that causes a slight damage to the tissue at the time of collection and recovers quickly, and to obtain cultured cells. Since methods for culturing stem cells are known, basically, cells are cultured according to the methods described herein and the known methods (for example, the methods described in Japanese Patent No. 5409359 and Japanese Patent No. 6041270). do it. The method for producing human skin-derived stem cells is also known techniques as described in the above-mentioned publication.
  • SAG is known by CAS No. 364590-63-6, and the chemical name is N-methyl-N '-(3-pyridinylbenzyl) -N'-(3-chlorobenzo [b] thiophene-2-carbonyl) It is a compound which is -1,4-diaminocyclohexane (N-Methyl-N '-(3-pyridinylbenzyl) -N'-(3-chlorobenzo [b] thiophene-2-carbonyl) -1,4-diaminocyclohexane).
  • SAG is a protein that activates sonic hedgehog (Shh) as disclosed in WO 2014-084085 pamphlet.
  • SAG 1.1 has the chemical name N-methyl-N '-(3- (4-benzonitrile) -4-methoxybenzyl) -N- (3-chlorobenzo [b] thiophene-2-carbonyl) -1, 4-Diaminocyclohexane (N-Methyl-N- (3- (4-benzonitrole) -4-methoxybenzyl) -N '-(3-chlorobenzo [b] thiophene-2-carbonyl) -1,4-diaminocyclohexane)) is there.
  • SAG is described in the document Sinha S, Chen JK. Nat Chem Biol. 2006 Jan; 2 (1): 29-30. And Chen JK, Taipale J, Young KE, Maiti T, Beachy PA. Proc Natl Acad Sci USA. 2002 Oct 29; 99 (22): 14071-6.
  • SAG 1.1 the documents Chen, W., Ren, X. R., Nelson, C. D., Barak, L. S., Chen, J. K., Beachy, P. A., de Sauvage, F. & Lefkowitz, R. J. (2004) Science 306, (5705) 2257-2260.
  • Palmorphamine (Purmorphamine) can be prepared by using (9-Cyclohexyl-N- [4- (4-morpholinyl) phenyl] -2- (1-naphthalenyloxy) -9H-purin-6-amine)) ((9- It is a general name of Cyclohexyl-N- [4- (4-morpholinyl) phenyl] -2- (1-naphthalenyloxy) -9H-purin-6-amine)).
  • Palmorphamine is a smoothed agonist also called purmorphamine and is described in Patents 6210881 and 5620821 (2 uM is added to the medium).
  • the Sonic hedgehog (SHH) protein is a known protein, and for example, in Japanese Patent No. 4900587, 200 to 400 ng / ml of SHH protein is added to a serum-free medium.
  • Serum-free medium The medium of the present invention is a serum-free medium, and may contain components in known media as appropriate. Also, for example, Japanese Patent No. 4385076 and Japanese Patent Application Laid-Open No. 2012-157263 disclose a medium for serum-free culturing of animal cells. Thus, the elements described in the known literature may be appropriately added to the medium of the present invention.
  • the culture medium of the present invention may be appropriately used as a basic culture medium.
  • a basic medium include MEM medium, Dulbecco's MEM (registered trademark) medium, MCDB 153, Ham's F12 (registered trademark) medium, McCoy 5A (registered trademark) medium, 199 medium air solution (registered trademark), RPMI 1640 (Registered trademark) medium, F-10 ham medium, MEM- ⁇ medium, DMEM / F12 medium, MCDB131, MCDB153 and MCDB201.
  • the pH of the culture medium is adjusted in the range of pH 6.8-7.8 when equilibrated in a 5% CO 2 environment, and more preferably pH 7.2 ( ⁇ 0.1).
  • the acidity of the basic medium may be appropriately adjusted using a pH adjuster such as a buffer (for example, sodium bicarbonate) or hydrochloric acid and sodium hydroxide.
  • the culture medium of the present invention may optionally contain various low molecular weight compounds.
  • the medium of the present invention preferably contains an antioxidant.
  • An example of an antioxidant is one or more components selected from the group consisting of melatonin (Melatonin), n-acetyl-L-cysteine, reduced glutathione and ascorbic acid.
  • the medium of the present invention includes phospholipids (eg, phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine) and fatty acids (eg, linoleic acid, oleic acid, linoleic acid, arachidonic acid, myristic acid, palmitoyl) Acid, palmitic acid, and stearic acid) may be included as appropriate. You may add to the culture medium of this invention.
  • phospholipids eg, phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine
  • fatty acids eg, linoleic acid, oleic acid, linoleic acid, arachidonic acid, myristic acid, palmitoyl
  • palmitic acid e.g., palmitic acid, and stearic acid
  • Examples of other components are transferrin, selenate, glucose, D-biotin, D-calcium pantothenate, choline chloride, folate, myoinositol, nicotinamide, p-aminobenzoic acid, pyridoxal hydrochloride, pyridoxi hydrochloride, riboflavin, Thiamine hydrochloride, vitamin B12, sodium pyruvate, thymidine, hypoxanthine, sodium selenite, streptomycin sulfate, penicillin G potassium salt, and phenol red.
  • the serum-free medium preferably further comprises a B-27 supplement.
  • a B-27 supplement When serum-free medium contains B-27 supplement, nestin mRNA is highly expressed as shown in the examples.
  • the nestin gene is a marker for neural stem cells.
  • B-27 supplement is a known element to be added to the culture medium, for example, in Patents 6185907 and 6137626.
  • the serum-free medium preferably further comprises a ROCK inhibitor.
  • ROCK inhibitors are defined as substances that inhibit the kinase activity of Rho-kinase (ROCK), for example Y-27632 (4-[(1R) -1-aminoethyl] -N-pyridin-4-ylcyclohexane- 1-carboxamide) or its dihydrochloride (for example, see Ishizaki et al., Mol. Pharmacol. 57, 976-983 (2000); Narumiya et al., Methods Enzymol.
  • ROCK Rho-kinase
  • Fasudil / HA1077 (1- (5-isoquinolinesulfonyl) homopiperazine) or its dihydrochloride (see, eg, Uenata et al., Nature 389: 990-994 (1997)), H-1152 ((S)-(+)-2 -Methyl-1-[(4-methyl-5-isoquinolinyl) sulfonyl] -hexahydro-1H-1,4-diazepine) or its dihydrochloride (eg Sasaki et al., Pharmacol. Ther.
  • ROCK inhibitors are generally used in the range of 100 nM to 50 ⁇ M.
  • the serum-free medium preferably further comprises EGF.
  • EGF epidermal growth factor
  • EGF is a growth factor selected from the epidermal growth factor (EGF) family.
  • EGF epidermal growth factor
  • the content of EGF in the medium is set to 0.5 to 200 ng / mL as the final concentration.
  • the GFAP gene is a marker for adult neural stem cells or astrocytes that is considered to be suitable for transplantation into spinal cord injury (Non-patent Documents 1, 20, 22).
  • the serum-free medium preferably further contains FGF2.
  • FGF fibroblast growth factor 2
  • FGF-2 fibroblast growth factor 2
  • FGF2 is added to the medium at a concentration of 0.1 to 50 ng / mL, more preferably 1 to 10 ng / mL, most preferably 5 ng / mL.
  • CD73 Ecto-5'-nucleotidase: Ecto-5'-nucleotidase
  • the serum-free medium preferably has a calcium ion concentration of not less than 0.03 mM and not more than 0.12 mM. By maintaining the calcium ion concentration within this range, stem cell differentiation can be suppressed.
  • Additives such as SAG, palmorphamine, sonic hedgehog (SHH) protein, B-27 supplement, ROCK inhibitor, EGF, FGF2 etc. should be added to the medium in an appropriate amount, taking into consideration the properties of those substances. It may be added. Since these substances are known to be added to the culture medium, the addition amount may be appropriately adjusted based on known materials. For example, it may be added to the culture medium to be 0.1 ng / mL or more and 20 ⁇ g / mL or less (or 0.2 ng / mL or more and 10 ⁇ g / mL or less). These may be appropriately adjusted and added according to the degree of purification and the required amount.
  • human skin-derived stem cells are cells obtained by treating collected human skin in the order of dispase treatment, trypsin treatment, and collagenase treatment.
  • Dispase treatment, trypsin treatment, and collagenase treatment are so-called enzyme treatments.
  • the enzyme treatment conditions may be an isotonic salt solution buffered to a physiologically acceptable pH, such as about 6 to 8, preferably about 7.2 to 7.6, such as PBS or Hanks balance salt in the case of trypsin.
  • the concentration may be sufficient for that, for example about 0.0001 to 5% w / v, preferably about 0.001% to 0.5% w / v.
  • the second aspect of the present invention also provides a method of producing a therapeutic agent for spinal cord injury.
  • the spinal cord is a central nerve that passes through the spine, and it is a therapeutic agent for spinal cord injury that is used to repair the nerve.
  • This method produces cultured cells using the method described above.
  • a therapeutic agent for a spinal cord injury disease which comprises one or more of a cultured cell, a culture medium obtained in a culture step, and a secretion (exosome etc.) obtained in a culture step as an active ingredient Agent).
  • the secretion (such as exosome) may be from culture supernatant. Also, as the medium, only the culture supernatant may be used.
  • the agent of the present invention may be produced by methods known to those skilled in the art.
  • the agent of the present invention can be manufactured as an oral preparation and a parenteral preparation, but is preferably a parenteral preparation.
  • a parenteral preparation may be a solution (aqueous solution, non-aqueous solution, suspension solution, emulsion solution, etc.) or a solid agent (powder filled formulation, freeze-dried preparation, etc.).
  • the agent of the present invention may be in the form of a sustained release preparation.
  • a dosage form in the case of using living cells as an agent a solution is preferable, and as a dosage form in the case of using a partial component of cells or whole dead cells, a solution or a solid agent can be selected.
  • the main components of the agent of the present invention are any one or more of cultured human skin-derived stem cells, a culture medium obtained in the culturing step, and secretions from cultured cells (exosome etc.).
  • the effective amount may be designed using the number of cells as an index.
  • Stem cells may be removed as the final agent.
  • the secretion may also be contained in the culture supernatant.
  • the dose and frequency of administration and the frequency of administration per dose vary depending on the subject, age, symptoms, etc. In general, an example of a daily dose of human skin-derived stem cell agent is 1 ⁇ 10 5 cells or more and 1 ⁇ 10 9 cells or more, preferably 1 ⁇ 10 6 cells or more, as one administration unit for intravenous injection.
  • an example of a daily dose of human skin-derived stem cells per kg body weight is 1 ⁇ 10 4 cells or more and 1 ⁇ 10 8 cells, preferably 1 ⁇ 10 5 cells or more and 5 ⁇ 10 7 cells, More preferably, it is 1 ⁇ 10 6 cells or more and 2 ⁇ 10 7 cells.
  • the agent of the present invention may be administered once every three days for a total of eight times, or daily administration may be continued for one week. It can also be used in combination for various frequency administration periods. In the case of significant response, where improvement of symptoms was observed at one time, it may be ended with only the first administration.
  • the agent of the present invention can be used in a wide variety of ways depending on the background of the disease or condition, and further, the number of transplanted cells in each administration can be arbitrarily set within the above-mentioned range .
  • the present invention also provides the use of the above-mentioned human skin-derived stem cells of the present invention for producing a therapeutic agent for spinal cord injury.
  • the invention also provides a method of treating spinal cord injury.
  • the method for treating various diseases comprises the step of administering an effective amount of the agent of the present invention produced as described above to a patient suffering from spinal cord injury.
  • the present invention can provide a cell culture method for safely increasing GDNF gene expression (mRNA) without employing a viral gene transfer method.
  • mRNA GDNF gene expression
  • the present invention can provide a cell culture method which also expresses various markers such as neural stem cell markers.
  • the present invention can provide a method for producing a therapeutic agent for spinal cord injury.
  • cells for treatment of spinal cord injury can be obtained from the skin which is easy to be collected among human tissues by the serum-free / feeder-free cell culture method.
  • a method of increasing the expression level of GDNF which is most essential for treatment of spinal cord injury by 10 times is found, together with this, nestin of neural stem cell marker is expressed to be effective for treatment of spinal cord injury. It was confirmed that cells that are highly effective in healing spinal cord injury, in which CDAP is also expressed, which expresses GFAP and prevents hyperalgesia caused by spinal cord injury, without gene transfer.
  • the cells obtained by the method of the present invention were confirmed to have a healing effect in a rat spinal cord injury model. Also, these cells were negative in oncogenic tests.
  • FIG. 1 is a graph replacing the drawing showing the effect of increasing the expression level of GDNF mRNA (Experimental Example 2).
  • FIG. 2 is a graph replaced with a drawing showing expression of Nestin mRNA (Experimental Example 3).
  • FIG. 3 is a graph replaced with a drawing showing expression of GFAP mRNA (Experimental Example 4).
  • FIG. 4 is a graph replaced with a drawing showing expression of CD73 (CD expression profile) (Experimental Example 5).
  • FIG. 5 is a graph replacing the drawing showing the effect (BBB score) (Experimental Example 6) in a rat spinal cord injury model.
  • FIG. 6 is a graph replacing a drawing showing evaluation of carcinogenicity.
  • the present invention is a tissue collecting process for collecting skin from human skin and disinfecting the tissue: Cell collection and seeding process: Cell proliferation process: Passage process: Differentiation induction process: Implantation / processing process: The present invention provides a method for obtaining cells that are highly effective for spinal cord injury through the above steps, and for providing cells to be directly administered, or for providing cells for a medical device combined with another base. Furthermore, a method of administering the post-culture medium component obtained in the above steps is provided.
  • u may mean micro.
  • tissue collection process The skin collected in this step is desirably located above the neck, and the skin on the back of the pinna is most preferable from the cosmetic point of view after collection.
  • the sex and age of the skin are not limited, and can be up to the 70s.
  • the following steps are all performed aseptically. Bacterial removal of the collected tissue is carried out with 70% ethanol water from which spores have been removed with a 0.1 um pore size removal filter.
  • the cut tissue is immersed in 10 ⁇ M Y-27632 in Dispase solution (PBS ( ⁇ )) at 4 ° C. overnight.
  • the tissue is then immersed in Trypsin solution (PBS ( ⁇ )) containing 10 uM Y-27632 at 37 ° C. for 20 minutes.
  • the tissue is immersed in collagenase solution (DMEM) and stirred at 37 ° C. for 1 to 2 hours with a stirrer to digest the skin. Remove the residue from the stirred suspension through a 100 um mesh.
  • the solution is centrifuged and washed three times with PBS (-) to remove the enzyme and obtain cells.
  • the cells are placed in Trypsin Inhibitor and allowed to stand for 10 minutes.
  • the cells are centrifuged to remove the inhibitor, and then suspended in a medium containing 10 uM Y-27632 and seeded in a culture dish.
  • Cell proliferation process The medium is not changed until the 6th day of seeding, and only the medium is added.
  • the subsequent medium exchange is carried out by centrifuging and recovering the cells floating in the conditioned medium, and the medium suspended in the new medium is returned to the culture system.
  • Passage process The cells are treated with 10 uM Y-27632 for 30 minutes or more, and the cells are recovered from the dish with EDTA and TrypLEx Express. Then place in Trypsin Inhibitor and let stand for 10 minutes. After centrifuging the cells to remove the Inhibitor, the cells are suspended in a medium containing 10 uM Y-27632 and seeded in the original dish of 4-5 times area. The cells are cultured in five 100 mm 2 dishes at passage 1 and when confluence is reached, three of them are used for transplantation or cryopreserved until transplantation. The remaining 2 sheets are further subcultured to 6 100 mm 2 dishes, and when confluence is reached, the process proceeds to the factor addition induction step.
  • the factor addition induction step is a step of carrying out drug treatment on required cells on the third day before transplantation, adding necessary factors, and culturing.
  • cells in which GDNF mRNA is highly expressed for example, cells in which 10 or more times normal culture is expressed are obtained.
  • this step produces CD13 negative cells (CD13 ( ⁇ )).
  • this step produces CD34 negative cells (CD34 ( ⁇ )).
  • this step produces CD45 negative cells (CD45 ( ⁇ )).
  • cells negative for CD90 can be obtained by this step (CD90 ( ⁇ )).
  • CD73-positive cells (CD73 (+)) are obtained by this step.
  • the cells that have reached the required number for treatment after each treatment are treated with 10 uM Y-27632 for 30 minutes or more, and the cells are recovered from dish with EDTA and TrypLEx Express. Then place in Trypsin Inhibitor and let stand for 10 minutes. After centrifuging the cells to remove the inhibitor, when transplanting by spinal cord puncture, it is used after suspension in a clinical fluid at a concentration of 10 7 cells / ml. The cells are subjected to processing with other carriers and cells.
  • the present invention also includes a range that can be appropriately adjusted by those skilled in the art using known techniques based on the following examples.
  • Cell collection seeding step 1 The cut tissue was immersed in 5 ml of a precooled Dispase solution (25 units / ml of PBS (-)) containing 10 uM Y-27632 overnight at 4 ° C.
  • Cell collection seeding step 2 After centrifuging this and removing the supernatant, it was immersed in TrypLE Express solution (5 ⁇ diluted PBS ( ⁇ )) containing 10 uM Y-27632 at 37 ° C. for 20 minutes.
  • Cell collection seeding step 3 The tissue was centrifuged to remove the supernatant, and then immersed in 20 ml of collagenase solution (GIBCO: type I) (1400 units / ml DMEM medium) and stirred with a stirrer at 37 ° C. for 1.5 hours.
  • GEBCO collagenase solution
  • Cell collection seeding step 4 The suspension after stirring was passed through a 100 um mesh to remove undigested residues. The solution was centrifuged and washed three times with PBS (-) to remove the enzyme, to obtain cells.
  • Cell collection seeding step 5 The cells were placed in 5 ml of soybean trypsin inhibitor (Soy bean Trypsin Inhibitor) (0.25% / PBS (-)) and allowed to stand for 10 minutes.
  • soybean trypsin inhibitor Soy bean Trypsin Inhibitor
  • Cell collection seeding process 6 The cells were centrifuged to remove the inhibitor, and then suspended in 10 uM Y-27632-added culture medium and seeded in culture dishes. From the size of the starting tissue (1 to 2 cm 2 ), it was seeded to all wells of one 6 well plate (CellBind: COSTAR).
  • Culture medium was prepared as follows. Specifically, insulin (10 mg / L), holo-transferrin (5.5 mg / L), selenium (6.7 ug / L), ethanolamine (2 mg / L) in MCDB 153 medium (Sigma: Stemline Keratinocyte Basal Medium) as a basic medium.
  • Vitamin C L-Ascorbic acid 2-phosphate semimagnesium salt
  • KGF 10 ug / L
  • lipid a mixture of fatty acid: arachidonic acid (20 ug / L), cholesterol (2.2 mg / L), DL- a-Tocopherol-acetate (700 ug / L), linoleic acid (100 ug / L), linolenic acid (100 ug / L), myristic acid (100 ug / L), oleic acid (100 ug / L), palmitoleic acid (100 ug / L) , Palmitic acid (100 ug / L), stearic acid (100 ug / L), Tween 80 (22 mg / L), Pluronic F-68 (1000 mg / L), EGF (20 ug / L), FGF (10 ug / L), Y- Prepared by adding 27632 (1 uM),
  • Cell proliferation step 1 In primary culture, conditioned culture was performed while adding medium at a rate of 1/3 volume without changing the medium until 7 days of culture. After that, culture was performed while changing the medium every two days. In this medium exchange, the old medium is centrifuged, 2/3 of its volume is discarded, the remaining 1/3 volume of medium and the floating cells precipitated by centrifugation are mixed, and 2/3 volume of new medium is further added to the culture dish. I'm back to. It took 10 days to reach 100% confluence.
  • Y-27632 was added at 10 uM concentration to the culture medium of the cultured cells and allowed to stand at 37 ° C. for 30 minutes. Next, the medium was completely removed, and 2 ml / well of 0.05% EDTA / PBS (-) was added and allowed to stand at 37 ° C. for 10 minutes. Furthermore, 0.5 ml / well of TrypLEx Express diluted 5-fold with PBS ( ⁇ ) was added, and the mixture was allowed to stand at 37 ° C. for 5 minutes. Next, the cells were detached from the wells by pipetting. To this, 2.5 ml / well of Trypsin Inhibitor (0.05% / PBS (-)) was added, and the recovered cells were centrifuged.
  • Trypsin Inhibitor 0.05% / PBS (-)
  • GDNF Glial cell-derived neurotrophic factor
  • VA Valproic acid
  • PDGF Platelete-derived growth factor
  • SAG or Purmorphamine or rh-Sonic Hedgehog (PeproTech)
  • BMP 4 Bone morphogenic protein 4
  • reaction solution Distilled water 11 ⁇ L SYBR® Green Realtime PCR Master Mix -Plus- 25 ⁇ L Plus solution 5 ⁇ L Primer 1 (10 ⁇ M) 2 ⁇ L Primer 2 (10 ⁇ M) 2 ⁇ L Sample solution 5 ⁇ L Total volume 50 ⁇ L
  • the reaction solution was placed in each well of a 96 well plate to carry out a PCR reaction.
  • the PCR cycle was a 3-step method, and 40 cycles of (1) to (2) were performed under the conditions of annealing 60 ° C / extension 72 ° C. Melting curve analysis was performed at the last step to confirm that no formation of primer dimer was observed.
  • FIG. 1 is a graph replaced with a drawing showing the measurement results of the expression level of GDNF mRNA in Example 2.
  • Example 1 7 (Expression change of Nestin mRNA by induction of neural differentiation) Kit for differentiation and identification of neural progenitor cells in 2 wells out of 3 wells that reached confluence in Example 1 7 using the culture medium (medium for NPC differentiation) of Neural Progenitor Cell Functional Identification Kit (R & D Systems SC082) It was cultured for a day. The other well was cultured in the maintenance medium of the invention for the same day. After extracting total RNA similarly to Example 2 on the 7th day, cDNA was prepared. Using this cDNA, the expression comparison of Nestin mRNA, which is a marker for neural stem cells, was measured by RT-PCR with GAPDH mRNA as the ground state.
  • the Primer of Nestin was Forward CGTTGGACAAGAGGTTGGAG (SEQ ID NO: 3) and Reverse TCCTGAAAGCTGAGGGAAG (SEQ ID NO: 4), and the expression chain length was 262 bp.
  • the same GAPDH primer as in Example 2 was used. The results are shown in FIG.
  • FIG. 2 is a graph replaced with a drawing showing the expression level of Nestin mRNA in Example 3.
  • the expression level of Nestin (Nestin mRNA / GAPDH mRNA) decreased to about 1/7 before differentiation.
  • the results showed that our growth medium maintained neural stem cells, and differentiation medium decreased neural stem cells.
  • Example 1 7 (Expression change of GFAP mRNA by induction of neural differentiation) Kit for differentiation and identification of neural progenitor cells in 2 wells out of 3 wells that reached confluence in Example 1 7 using the culture medium (medium for NPC differentiation) of Neural Progenitor Cell Functional Identification Kit (R & D Systems SC082) It was cultured for a day. The other well was cultured in the maintenance medium of the invention for the same day. After extracting total RNA similarly to Example 2 on the 7th day, cDNA was prepared. Using this cDNA, the expression comparison of GFAP mRNA, which is a marker of Astrocytes or Adult neural stem cells, was measured by RT-PCR with GAPDH mRNA as the basal state.
  • GFAP mRNA which is a marker of Astrocytes or Adult neural stem cells
  • the GFAP Primer was Forward: ACATCGAGATCGCCCACCTAC (SEQ ID NO: 5) and Reverse: ACATCACATCCTTGTGCTCC (SEQ ID NO: 6), and the expression chain length was 219 bp.
  • the same GAPDH primer as in Example 2 was used. The results are shown in FIG.
  • FIG. 3 is a graph replaced with a drawing showing the expression level of GFAP mRNA in Example 4.
  • the expression level of GFAP (GFAP mRNA / GAPDH mRNA) slightly increased when it was changed to the differentiation medium, compared to before differentiation.
  • the results show that the inventors' growth media maintain neural stem cells and also maintain the expression of GFAP in them.
  • FIG. 4 is a graph replaced with a drawing showing expression of CD73 (CD expression profile) in Example 5.
  • CD13, CD34 and CD45 were negative, indicating that the cells were non-hematologic cells.
  • CD90 was also negative, indicating that it is not human skin-derived mesenchymal stem cells.
  • the strong expression of CD73 was the most distinctive feature of CD in this cell.
  • the therapeutic effect of spinal cord injury was measured by the BBB Score method in which the movement of the animal was observed with a video device and scored. The results are shown in the figure.
  • the horizontal axis shows the number of days after cell transplantation, and the vertical axis shows BBB Score.
  • the solid line shows the result of transplantation of the above cells, the dotted line shows the result of the control, * shows that the result of the significant difference test by Student T test is P ⁇ 0.05, ** is P ⁇ 0.01 and *** is P ⁇ 0.001.
  • the cell transplantation group showed a statistically significant effect (P ⁇ 0.05) with a 2-point difference in BBB Score compared to the control group.
  • the cell transplantation group showed a statistically significant effect (P ⁇ 0.001) with a 4-point difference in BBB Score compared to the control group.
  • P ⁇ 0.001 a 4-point difference in BBB Score compared to the control group.
  • the significant difference was P ⁇ 0.05, but the present invention was more dominant with P ⁇ 0.001 at the same time. This result shows that the cells obtained by the present invention are very effective in the treatment of spinal cord injury.
  • mice (6 weeks old, rabbit, Central Research Institute for Experimental Animals) having NOD / Shi-scid, IL-2R ⁇ null (registered trademark) strain were used. After one week of acclimatization period, using a universal grouping system (Visions Co., Ltd.), groups were divided into three groups with the same average weight value as possible, with the right flank of each group of mice.
  • Example 2 In the same manner as in Example 1 (cultured as cell A), a culture solution as negative control cells, and HeLa S3 (DS Pharma Biomedical Co., Ltd.) as positive control cells were each syringe (Myjector (registered trademark), Needle specifications (27 G) were transplanted (both in 0.2 mL / individual).
  • each group be cell A group (10 individuals), culture fluid group (10 individuals), and HeLa S3 group (5 individuals).
  • For cell A prepare 3 lines of cells cultured in the same manner as in Example 1, suspend each at 1 ⁇ 10 7 cells / mL in ⁇ -MEM medium, and divide into 2 ml cryotubes so as not to contain air. I ordered.
  • mice in each group are bred 13 weeks after transplantation in accordance with the “Working standard for breeding and management of experimental animals at the Central Research Institute for Experimental Animals of the Public Interest Foundation”, general condition observation, weight measurement and observation of nodules -Size measurement was performed once a week.
  • mice in the HeLaS3 group have 1/10 weight of the body weight (the specific gravity is 1 and the volume is calculated from the volume) during the observation period. Since it was confirmed that it was exceeded, observation was ended as a humanitarian endpoint at weeks 5 to 8, and euthanasia was given.
  • the mice were euthanized by total bleeding under isoflurane anesthesia, and the chest and abdominal organs were observed.
  • the skin at the transplantation site was collected including subcutaneous tissue and fixed with 10% neutral buffered formalin solution. Formalin-fixed specimens were paraffin-embedded, sliced, HE staining, anti-HLA (Human Leucocyte Antigen) immunostaining, and observed with a light microscope in a conventional manner.
  • HLA Human Leucocyte Antigen
  • FIG. 6 is a graph replacing a drawing showing evaluation of carcinogenicity.
  • nodule formation was observed in 100% (5 of 5) individuals at 3 weeks after transplantation, but no nodule formation was observed in the culture fluid group and cell group A until the end of observation
  • the Nodules were formed in all cases in the HeLa S3 group.
  • the nodule was considered to be a tumor derived from HeLa S3 cells, as it showed continuous volume increase and showed anti-HLA reaction together with the histological image of poorly differentiated carcinoma.
  • histopathological examination confirmed fiber proliferation showing anti-HLA positive in 6/10 cases, and it was judged that the transplanted cells remained.
  • the present invention can be utilized in the pharmaceutical industry.
  • Sequence number 1 Primer sequence number 2: Primer sequence number 3: Primer sequence number 4: Primer sequence number 5: Primer sequence number 6: Primer

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Dermatology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Ophthalmology & Optometry (AREA)
  • Physics & Mathematics (AREA)
  • Toxicology (AREA)
  • Psychology (AREA)
  • Plant Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)

Abstract

[Problem] To provide a cell culture method which can achieve the increase in safe expression of GDNF gene (mRNA) without needing to employ a gene transfer method using a virus. [Solution] A method for producing a cultured cell containing mRNA for glial cell line derived neurotrophic factor (GDNF), the method including a culture step of culturing a human skin-derived stem cell in a serum-free culture medium containing at least one component selected from SAG, purmorphamine and sonic hedgehog (SHH) protein, wherein the serum-free culture medium may additionally contain a B-27 supplement, a ROCK inhibitor, EGF and FGF2.

Description

培養細胞の製造方法,脊髄損傷疾患の治療剤の製造方法Method of producing cultured cells, method of producing therapeutic agent for spinal cord injury disease
この発明は,ヒト皮膚組織から得られる細胞を培養して得られる,脊髄損傷疾患群を治療するための細胞の製造方法に関し,また,前記方法により得られる細胞または培養培地またはExosome等の分泌物を含有する脊髄損傷疾患群を治療する医療薬の製造方法に関する。 The present invention relates to a method for producing cells obtained by culturing cells obtained from human skin tissue for treating a spinal cord injury disease group, and cells obtained by the above-mentioned method or a secretion such as culture medium or Exosome. The present invention relates to a method for producing a medical drug for treating a spinal cord injury disease group containing
 脊髄損傷(Spinal Cord Injury)は,主として脊柱に強い外力が加えられることにより脊椎を損壊し,脊髄に損傷をうける病態である。また,脊髄腫瘍やヘルニアや頸椎脊髄症など内的原因によっても類似の障害が発生する。脊髄損傷の推定患者数は,日本では10万人である。脊髄損傷の原因の大半は交通事や高所からの落下等による外傷である。 Spinal cord injury is a condition in which the spine is damaged mainly by applying a strong external force to the spinal column, and the spinal cord is damaged. Similar disorders also occur with internal causes such as spinal cord tumors, hernias and cervical spinal myelopathy. The estimated number of patients with spinal cord injury is 100,000 in Japan. Most of the causes of spinal cord injuries are injuries due to traffic or falling from high places.
 脊髄を含む中枢神経系は,末梢神経と異なり,一度損傷すると修復・再生されることは無い。破壊された脊髄を再生し,再び機能を取り戻すことは全世界の脊損者の願いであり,様々な分野で研究が進められている。 Unlike peripheral nerves, the central nervous system including the spinal cord can not be repaired or regenerated once it is damaged. Regeneration of the destroyed spinal cord and regaining its function are the wishes of spinal cord all over the world, and research is underway in various fields.
 現在受傷直後では,米国において,受傷直後,48~72時間以内であれば,大量にステロイド剤の投与が行われる。日本の関西医科大学において,やはり受傷直後の患者に対し,自分の骨髄液を培養して脊髄に注入したり,日本の慶応大学などのグループが事故などで脊髄を損傷して78時間以内に,肝細胞増殖因子(HFG)を投与するなどの試みが行われている。 At present, immediately after injury, in the United States, a large amount of steroid agent administration is performed within 48 to 72 hours immediately after injury. For patients immediately after injury at Kansai Medical University in Japan, their bone marrow fluid is cultured and injected into the spinal cord, and groups such as Keio University in Japan have injured the spinal cord within 78 hours after an accident, etc. Attempts have been made to administer hepatocyte growth factor (HFG) and the like.
 慢性期になった場合は,リハビリテーションが行われる。しかし,脊髄損傷のリハビリテーションは失われた機能を回復させることではない。神経が再生しない以上,それは不可能だからである。リハビリの目的は,「残された機能をいかにして使い,ADL(日常生活動作)を可能にするか」という点にある。更に慢性期になると褥瘡や尿路感染症などの合併症が発生し,介護は困難を極める。 In the chronic phase, rehabilitation is performed. However, rehabilitation of spinal cord injury is not to restore lost function. That is because it is impossible if the nerve does not regenerate. The purpose of rehabilitation is "how to use the remaining functions and enable ADL (Daily life movement)". Furthermore, complications such as hemorrhoids and urinary tract infections occur in the chronic stage, and care becomes extremely difficult.
 その慢性期でも有効な治療法として現在最も有望視されているのが,骨髄や神経の幹細胞を用いた神経再生の試みである。動物実験では部分的な効果が報告されているが,人体に応用し治療に役立つには未だ基礎研究の段階であり,研究の強力な推進が望まれている。主として人工多能性幹細胞,胚性幹細胞の使用が研究されているが,解決すべき課題も多い。例として,人工多能性幹細胞(iPS)を用いた場合,免疫反応は回避できると考えられるものの,腫瘍となる可能性が指摘されている。また,胚性幹細胞については,免疫反応や腫瘍化の問題に加え,倫理面での問題が指摘されている。 Even in the chronic phase, attempts at nerve regeneration using bone marrow and neural stem cells are currently most promising as effective treatments. Although partial effects have been reported in animal experiments, it is still a basic research stage to be applied to the human body and to be useful for treatment, and strong promotion of research is desired. Although mainly the use of induced pluripotent stem cells and embryonic stem cells has been studied, there are also many issues to be solved. As an example, when induced pluripotent stem cells (iPS) are used, although it is thought that the immune response can be avoided, the possibility of becoming a tumor is pointed out. In addition to the problems of immune response and tumorigenesis, there are also ethical issues with embryonic stem cells.
 しかし倫理面と免疫拒絶の問題をクリアーした遺伝子導入から造られたiPS細胞を用いた脊損動物への投与では脊髄内に腫瘍ができるとの報告が日本から2010年に発表されている(非特許文献1)。 However, it has been announced in 2010 from Japan that a tumor can be generated in the spinal cord by administration to spinal cord animals using iPS cells produced from gene transfer that has cleared the issue of ethics and immune rejection (non-patented in 2010) Literature 1).
 2005年現在,唯一臨床治療として行われているのが,中国の北京首都医科大学において,鼻臭覚粘膜細胞(OEG)を注入することで脊髄の再生を図るというものである。しかし同大からは長期にわたる治療効果の検証において,世界の研究者を納得させるデータの提出が無く,激しい疼痛やOEGの入手先(中絶胎児から採取)の問題や他家移植の問題が残されている。2014年に発表された自己の鼻粘膜を移植する日本の臨床試験でも劇的効果(歩行可能例)は報告されていない。 As of 2005, the only clinical treatment that has been performed is that, at Beijing Metropolitan Medical University in China, regeneration of the spinal cord is intended by injecting nasal olfactory membrane cells (OEG). However, from the same university in the long-term examination of the therapeutic effect, there is no submission of data convincing researchers in the world, leaving the problem of intense pain, the source of OEG (from abortion fetus) and the problem of cross transplantation ing. No dramatic effects (walkable cases) have been reported in Japanese clinical trials for transplanting the own nasal mucosa, which were announced in 2014.
 2010年10月,アメリカのジェロン社が脊髄損傷の患者4人に対しES細胞を使用した臨床試験を開始したが,2011年11月に撤退を発表している。この原因は不明であるが,臨床効果が乏しかったか,或は免疫的な拒絶反応が現れたか,または脊髄内にTeratomaが発生した可能性が高い。
慶應大学の中村雅也准教授らのグループが京都市で開かれた日本再生医療学会で,脊髄損傷の患者に対するiPS細胞の臨床研究を2017年度に始める計画を発表した。対象は,事故から2~4週間後で,患部の炎症が収まり傷口が固まり始める前の患者であり,慢性期の患者は適応外である。
In October 2010, Geron in the US launched a clinical trial using ES cells in four patients with spinal cord injury, but announced withdrawal in November 2011. Although the cause is unknown, it is likely that the clinical effect was poor or immunologic rejection occurred or that Teratoma occurred in the spinal cord.
A group of Associate Professors Masaya Nakamura of Keio University announced a plan to start clinical research on iPS cells for patients with spinal cord injury in 2017 at the Japan Society for Regenerative Medicine held in Kyoto City. Subjects are patients 2 to 4 weeks after the accident, before the inflammation in the affected area subsides and the wound starts to harden, and patients in the chronic phase are not indicated.
 このように幹細胞治療が注目 されているが,上市済の再生医療製品は,米国で幹細胞に よるものは1品目,EUで 前進治療医薬品(Advanced Therapy Medicinal Product)として1品目のみである。しかしながら, 脊髄損傷に対して承認された再生医療製品は存在しない現状である。 As such, although stem cell therapy has attracted attention, there is only one marketed regenerative medicine product in the United States that uses stem cells, and one in the EU as Advanced Therapy Medicinal Products. However, there is currently no regenerative medicine product approved for spinal cord injury.
 また,ヒト皮膚由来幹細胞(Mesenchymal stem cells)は,移植後に神経細胞に誘導し難く,軟骨細胞や骨細胞になってしまう可能性が高く,脳由来神経幹細胞は治療効果が低い等の欠点を有する(非特許文献13)。 In addition, human skin-derived stem cells (Mesenchymal stem cells) are difficult to induce in neurons after transplantation and are likely to become chondrocytes and osteocytes, and brain-derived neural stem cells have disadvantages such as low therapeutic effect (Non-Patent Document 13).
   一方,皮膚からの前駆細胞は脳からの神経幹細胞よりも脊損での治療効果が高いことが報告されている(非特許文献14)。 On the other hand, it has been reported that precursor cells from the skin are more effective in treating spinal cord injury than neural stem cells from the brain (Non-patent Document 14).
  更に最近,井上&熊谷らは,ヒト表皮keratinocytesにも脊髄損傷への治癒効果があることも報告している(非特許文献2)。 Furthermore, Inoue & Kumagaya et al. Also recently reported that human epidermal keratinocytes also have a healing effect on spinal cord injury (Non-patent Document 2).
 しかしながら,未だに脊髄損傷に対し,治療効果のマーカーを特定し,それらを発現した細胞の培養調製法は存在しない。 However, for spinal cord injury, there are still no markers of therapeutic effect, and there is no culture preparation method for cells expressing them.
   一方,サイトカインとしてはグリア細胞株由来神経栄養因子(Glial cell line-derived neurotrophic factor:GDNF)が脊髄損傷に効果が最も明確に示されているものである(非特許文献4-8)。 On the other hand, as cytokines, glial cell line-derived neurotrophic factor (GDNF) is the one most clearly shown to be effective in spinal cord injury (Non-patent Document 4-8).
 そこで,更に脊髄損傷の治癒効果を上げるものを造ろうとする試みは治癒効果の期待される細胞に更にGDNFの遺伝子を導入する試みである(非特許文献8-10)。 遺伝子を導入する細胞としては嗅覚受容細胞(Olfactory ensheathing cells)や,シュワン細胞(Schwann cells)等が使用されており,この細胞自身でも治癒効果を持つので,更なる治療効果の増大がある。 Therefore, an attempt to further create a healing effect of spinal cord injury is an attempt to further introduce the gene of GDNF into cells expected to have a healing effect (Non-patent Document 8-10). As cells into which genes are introduced, Olfactory ensheathing cells, Schwann cells, etc. are used, and since these cells themselves have a healing effect, there is a further increase in the therapeutic effect.
 しかし,この方法の欠点はウィルスを用いて遺伝子導入するため,生体に移植した時に癌化する可能性が高い。 However, the drawback of this method is that the gene is transferred using a virus, so there is a high possibility of becoming cancerous when transplanted into a living body.
 逆に,GDNFの蛋白そのものは不安定で取扱いが困難である(非特許文献15)。 Conversely, GDNF protein itself is unstable and difficult to handle (Non-patent Document 15).
 また,GDNFは脊髄損傷の回復に必要な内因性因子としても報告されている(非特許文献16)。 GDNF has also been reported as an intrinsic factor required for the recovery of spinal cord injury (Non-patent Document 16).
 さらに,GDNFは脊髄損傷患者を悩ます異痛症を抑制する効果も報告されている(非特許文献17-18)。 Furthermore, GDNF has also been reported to be effective in suppressing allodynia that afflicts patients with spinal cord injury (Non-patent Literature 17-18).
 脊髄損傷の治療の多くは慢性期に入った患者に対するものであり,慢性期にも効果のある治療法が最も望まれる。幸い,GDNFは慢性期の脊髄損傷にも効果が報告されている(非特許文献19)。 Most of the treatments for spinal cord injuries are for patients who have entered the chronic phase, and treatments that are effective in the chronic phase are most desirable. Fortunately, GDNF has also been reported to be effective in chronic spinal cord injury (Non-patent Document 19).
 したがって,移植する細胞にGDNFを発現させることは,脊髄損傷の治療効果を画期的に高めることにつながる(非特許文献20)。 Therefore, expressing GDNF in cells to be transplanted leads to a breakthrough in the therapeutic effect of spinal cord injury (Non-patent Document 20).
特開2012-29684号公報JP, 2012-29684, A
 本発明は,ウィルスによる遺伝子導入法を採用せずに,安全なGDNF遺伝子発現(mRNA)の増加を行う細胞培養方法を提供することを目的とする。 An object of the present invention is to provide a cell culture method for safely increasing GDNF gene expression (mRNA) without adopting viral gene transfer method.
 本発明は,神経幹細胞のマーカーなど種々のマーカーをも発現する細胞培養方法を提供することを目的とする。 An object of the present invention is to provide a cell culture method which also expresses various markers such as markers of neural stem cells.
 本発明は,脊髄損傷疾患の治療剤の製造方法を提供することを目的とする。 An object of the present invention is to provide a method for producing a therapeutic agent for spinal cord injury.
 本発明は,基本的には,ヒト皮膚由来幹細胞を,SAG,パルモルファミン(Purmorphamine)及びソニック・ヘッジホッグ (Sonic hedgehog, SHH)タンパク質のいずれか1種又は2種以上を含む無血清培地で培養することで,グリア細胞株由来神経栄養因子(GDNF)mRNAを高発現(例えば,通常培養の10倍以上発現)させることができるという実施例による知見に基づく。 Basically, the present invention is a serum-free medium containing human skin-derived stem cells and any one or more of SAG, palmorphamine and sonic hedgehog (SHH) proteins. It is based on the findings from the example that by culturing, glial cell line-derived neurotrophic factor (GDNF) mRNA can be highly expressed (for example, 10 times or more of that in normal culture).
 本発明の第1の側面は,グリア細胞株由来神経栄養因子(GDNF)mRNAを含む培養細胞の製造方法に関する。この方法は,ヒト皮膚由来幹細胞を,SAG,パルモルファミン(Purmorphamine)及びソニック・ヘッジホッグ (Sonic hedgehog, SHH)タンパク質のいずれか1種又は2種以上を含む無血清培地で培養する培養工程を含む。この方法は,グリア細胞株由来神経栄養因子(GDNF)mRNAが高発現する。 A first aspect of the present invention relates to a method for producing a cultured cell containing glial cell line-derived neurotrophic factor (GDNF) mRNA. This method involves culturing a human skin-derived stem cell in a serum-free medium containing one or more of SAG, palmorphamine (Purmorphamine) and Sonic hedgehog (SHH) proteins. Including. This method highly expresses glial cell line-derived neurotrophic factor (GDNF) mRNA.
 ヒト皮膚由来幹細胞は,患者本人の皮膚細胞に由来するものが好ましい。特に,採取時に組織への損傷が軽度で回復が早い皮膚組織からの細胞を採取し,培養細胞を得ることが好ましい。幹細胞の培養方法は公知であるから,基本的には本明細書に記載された方法と,公知の方法(例えば特許第5409359号公報や特許第6041270号公報に記載された方法)に従って細胞を培養すればよい。ヒト皮膚由来幹細胞の製造方法も,上記の公報に記載されているほか公知技術である。 Human skin-derived stem cells are preferably derived from skin cells of the patient himself. In particular, it is preferable to collect cells from skin tissue that causes a slight damage to the tissue at the time of collection and recovers quickly, and to obtain cultured cells. Since methods for culturing stem cells are known, basically, cells are cultured according to the methods described herein and the known methods (for example, the methods described in Japanese Patent No. 5409359 and Japanese Patent No. 6041270). do it. The method for producing human skin-derived stem cells is also known techniques as described in the above-mentioned publication.
 SAGは,CAS番号364590-63-6で知られ,化学物質名がN-メチル-N’-(3-ピリジニルベンジル)-N’-(3-クロロベンゾ[b]チオフェン-2-カルボニル)-1,4-ジアミノシクロヘキサン(N-Methyl-N’-(3-pyridinylbenzyl)-N’-(3-chlorobenzo[b]thiophene-2-carbonyl)-1,4-diaminocyclohexane)である化合物である。 SAG is known by CAS No. 364590-63-6, and the chemical name is N-methyl-N '-(3-pyridinylbenzyl) -N'-(3-chlorobenzo [b] thiophene-2-carbonyl) It is a compound which is -1,4-diaminocyclohexane (N-Methyl-N '-(3-pyridinylbenzyl) -N'-(3-chlorobenzo [b] thiophene-2-carbonyl) -1,4-diaminocyclohexane).
 SAGは,国際公開WO2014-084085号パンフレットに開示される通り,ソニックヘッジホッグ(Shh)を活性化させるタンパク質である。SAG1.1は,化学物質名がN-メチル-N’-(3-(4-ベンゾニトリル)-4-メトキシベンジル)-N-(3-クロロベンゾ[b]チオフェン-2-カルボニル)-1,4-ジアミノシクロヘキサン(N-Methyl-N-(3-(4-benzonitrile)-4-methoxybenzyl)-N’-(3-chlorobenzo[b]thiophene-2-carbonyl)-1,4-diaminocyclohexane))である。SAGについては文献Sinha S, Chen JK. Nat Chem Biol. 2006 Jan;2(1):29-30.及びChen JK, Taipale J, Young KE, Maiti T, Beachy PA. Proc Natl Acad Sci U S A. 2002 Oct 29;99(22):14071-6.に詳しい。同様に,SAG1.1については文献Chen, W., Ren, X. R., Nelson, C. D., Barak, L. S., Chen, J. K., Beachy, P. A.,de Sauvage, F. & Lefkowitz, R. J. (2004) Science 306,(5705) 2257-2260に詳しい。 SAG is a protein that activates sonic hedgehog (Shh) as disclosed in WO 2014-084085 pamphlet. SAG 1.1 has the chemical name N-methyl-N '-(3- (4-benzonitrile) -4-methoxybenzyl) -N- (3-chlorobenzo [b] thiophene-2-carbonyl) -1, 4-Diaminocyclohexane (N-Methyl-N- (3- (4-benzonitrole) -4-methoxybenzyl) -N '-(3-chlorobenzo [b] thiophene-2-carbonyl) -1,4-diaminocyclohexane)) is there. SAG is described in the document Sinha S, Chen JK. Nat Chem Biol. 2006 Jan; 2 (1): 29-30. And Chen JK, Taipale J, Young KE, Maiti T, Beachy PA. Proc Natl Acad Sci USA. 2002 Oct 29; 99 (22): 14071-6. Similarly, for SAG 1.1, the documents Chen, W., Ren, X. R., Nelson, C. D., Barak, L. S., Chen, J. K., Beachy, P. A., de Sauvage, F. & Lefkowitz, R. J. (2004) Science 306, (5705) 2257-2260.
 パルモルファミン(Purmorphamine)は,(9-シクロヘキシル-N- [4-(4-モルホリニル)フェニル] -2-(1-ナフタレニルオキシ)-9H-プリン-6-アミン))((9-Cyclohexyl-N-[4-(4-morpholinyl)phenyl]-2-(1-naphthalenyloxy)-9H-purin-6-amine))の一般名である。パルモルファミンは,プルモルファミンともよばれるスムーズンドアゴニストであり,特許6210881号公報,及び特許5620821号公報(2uMが培地に添加されている)に記載されている。 Palmorphamine (Purmorphamine) can be prepared by using (9-Cyclohexyl-N- [4- (4-morpholinyl) phenyl] -2- (1-naphthalenyloxy) -9H-purin-6-amine)) ((9- It is a general name of Cyclohexyl-N- [4- (4-morpholinyl) phenyl] -2- (1-naphthalenyloxy) -9H-purin-6-amine)). Palmorphamine is a smoothed agonist also called purmorphamine and is described in Patents 6210881 and 5620821 (2 uM is added to the medium).
 ソニック・ヘッジホッグ (Sonic hedgehog, SHH)タンパク質は,公知のタンパク質であり,例えば特許4900587号公報では,200~400ng/mlのSHHタンパク質が無血清培地に添加されている。 The Sonic hedgehog (SHH) protein is a known protein, and for example, in Japanese Patent No. 4900587, 200 to 400 ng / ml of SHH protein is added to a serum-free medium.
 無血清培地
 本発明の培地は,無血清培地であり,公知の培地における成分を適宜含んでもよい。また,例えば,特許第4385076号公報及び特開2012-157263号公報には動物細胞を無血清培養するための培地が開示されている。このように公知の文献に記載される要素を適宜本発明の培地に添加してもよい。
Serum-free medium The medium of the present invention is a serum-free medium, and may contain components in known media as appropriate. Also, for example, Japanese Patent No. 4385076 and Japanese Patent Application Laid-Open No. 2012-157263 disclose a medium for serum-free culturing of animal cells. Thus, the elements described in the known literature may be appropriately added to the medium of the present invention.
 本発明の培地は,基本培地として公知のものを適宜用いてもよい。そのような基本培地の例は,MEM培地,ダルベッコMEM(登録商標)培地,MCDB153,ハムF12(登録商標)培地,マッコイ5A(登録商標)培地,199培地ア―ル液(登録商標),RPMI1640(登録商標)培地,F―10ハム培地,MEM-α培地,DMEM/F12培地,MCDB131,MCDB153及びMCDB201である。 The culture medium of the present invention may be appropriately used as a basic culture medium. Examples of such a basic medium include MEM medium, Dulbecco's MEM (registered trademark) medium, MCDB 153, Ham's F12 (registered trademark) medium, McCoy 5A (registered trademark) medium, 199 medium air solution (registered trademark), RPMI 1640 (Registered trademark) medium, F-10 ham medium, MEM-α medium, DMEM / F12 medium, MCDB131, MCDB153 and MCDB201.
 培地のpHは,5%濃度CO環境で平衡化された際にpH6.8-7.8の範囲で調整され,pH7.2(±0.1)であることがより好ましい。緩衝材(例えば,重炭酸ナトリウム)や塩酸及び水酸化ナトリウムといったpH調整剤を用いて,基本培地の酸性度を適宜調整してもよい。 The pH of the culture medium is adjusted in the range of pH 6.8-7.8 when equilibrated in a 5% CO 2 environment, and more preferably pH 7.2 (± 0.1). The acidity of the basic medium may be appropriately adjusted using a pH adjuster such as a buffer (for example, sodium bicarbonate) or hydrochloric acid and sodium hydroxide.
 本発明の培地は,様々な低分子化合物を適宜含んでもよい。たとえば,本発明の培地は,抗酸化剤を含むことが好ましい。抗酸化剤の例は,メラトニン(Melatonin),n-アセチル-L-システイン,還元型グルタチオン及びアスコルビン酸からなる群から選択される1または2以上の成分である。本発明の培地は,リン脂質(例えば,フォスファチジルセリン,フォスファチジルエタノールアミン,及びフォスファチジルコリンなど)や脂肪酸(例えば,リノール酸,オレイン酸,リノレイン酸,アラキドン酸,ミリスチン酸,パルミトイル酸,パルミチン酸,及びステアリン酸)を適宜含んでもよい。本発明の培地に添加してもよい。他の成分の例は,トランスフェリン,セレン酸塩,グルコース,D-ビオチン,D-パントテン酸カルシウム,塩化コリン,葉酸,ミオイノシトール,ニコチンアミド,p-アミノ安息香酸,ピリドキサール塩酸,塩酸ピリドキシ,リボフラビン,塩酸チアミン,ビタミンB12,ピルビン酸ナトリウム,チミジン,ヒポキサンチン,亜セレン酸ナトリウム,硫酸ストレプトマイシン,ペニシリンGカリウム塩,及びフェノールレッドなどである。 The culture medium of the present invention may optionally contain various low molecular weight compounds. For example, the medium of the present invention preferably contains an antioxidant. An example of an antioxidant is one or more components selected from the group consisting of melatonin (Melatonin), n-acetyl-L-cysteine, reduced glutathione and ascorbic acid. The medium of the present invention includes phospholipids (eg, phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine) and fatty acids (eg, linoleic acid, oleic acid, linoleic acid, arachidonic acid, myristic acid, palmitoyl) Acid, palmitic acid, and stearic acid) may be included as appropriate. You may add to the culture medium of this invention. Examples of other components are transferrin, selenate, glucose, D-biotin, D-calcium pantothenate, choline chloride, folate, myoinositol, nicotinamide, p-aminobenzoic acid, pyridoxal hydrochloride, pyridoxi hydrochloride, riboflavin, Thiamine hydrochloride, vitamin B12, sodium pyruvate, thymidine, hypoxanthine, sodium selenite, streptomycin sulfate, penicillin G potassium salt, and phenol red.
 上記の培養細胞の製造方法は,無血清培地が,B-27サプリメントをさらに含むことが好ましい。無血清培地がB-27サプリメントを含むと,実施例において示された通り,ネスチンmRNAが高発現する。ネスチン遺伝子は,神経幹細胞のマーカーである。B-27サプリメントは,例えば,特許6185907号公報,及び特許6137626号公報において培地に添加されるように公知の要素である。 In the method for producing cultured cells described above, the serum-free medium preferably further comprises a B-27 supplement. When serum-free medium contains B-27 supplement, nestin mRNA is highly expressed as shown in the examples. The nestin gene is a marker for neural stem cells. B-27 supplement is a known element to be added to the culture medium, for example, in Patents 6185907 and 6137626.
 上記の培養細胞の製造方法は,無血清培地が,ROCK阻害剤をさらに含むことが好ましい。無血清培地がROCK阻害剤を含むと,幹細胞の安定性が維持される。ROCK阻害剤は,Rho-キナーゼ(ROCK)のキナーゼ活性を阻害する物質として定義され,例えば,Y-27632(4-[(1R)-1-アミノエチル]-N-ピリジン-4-イルシクロヘキサン-1-カルボキサミド)又はその2塩酸塩(例えば,Ishizaki et al., Mol. Pharmacol. 57, 976-983 (2000);Narumiya et al., Methods Enzymol. 325,273-284 (2000)参照),Fasudil/HA1077(1-(5-イソキノリンスルホニル)ホモピペラジン)又はその2塩酸塩(例えば,Uenata et al., Nature 389: 990-994 (1997)参照),H-1152((S)-(+)-2-メチル-1-[(4-メチル-5-イソキノリニル)スルホニル]-ヘキサヒドロ-1H-1,4-ジアゼピン)又はその2塩酸塩(例えば,Sasaki et al., Pharmacol. Ther. 93: 225-232 (2002)参照),Wf-536((+)-(R)-4-(1-アミノエチル)-N-(4-ピリジル)ベンズアミド1塩酸塩)(例えば,Nakajima et al., Cancer Chemother. Pharmacol. 52(4): 319-324 (2003)参照)及びそれらの誘導体,並びにROCKに対するアンチセンス核酸,RNA干渉誘導性核酸(例えば,siRNA),ドミナントネガティブ変異体,及びそれらの発現ベクターが挙げられる。また,ROCK阻害剤としては他の低分子化合物も知られているので,本発明においてはこのような化合物又はそれらの誘導体も使用できる(例えば,米国特許出願公開第20050209261号,同第20050192304号,同第20040014755号,同第20040002508号,同第20040002507号,同第20030125344号,同第20030087919号,及び国際公開第2003/062227号,同第2003/059913号,同第2003/062225号,同第2002/076976号,同第2004/039796号参照)。本発明では,少なくとも1種のROCK阻害剤が使用され得る。ROCK阻害剤は,一般的に100nM~50μMの範囲で用いられている。 In the method for producing cultured cells described above, the serum-free medium preferably further comprises a ROCK inhibitor. When serum-free medium contains ROCK inhibitor, stem cell stability is maintained. ROCK inhibitors are defined as substances that inhibit the kinase activity of Rho-kinase (ROCK), for example Y-27632 (4-[(1R) -1-aminoethyl] -N-pyridin-4-ylcyclohexane- 1-carboxamide) or its dihydrochloride (for example, see Ishizaki et al., Mol. Pharmacol. 57, 976-983 (2000); Narumiya et al., Methods Enzymol. 325, 273-284 (2000)), Fasudil / HA1077 (1- (5-isoquinolinesulfonyl) homopiperazine) or its dihydrochloride (see, eg, Uenata et al., Nature 389: 990-994 (1997)), H-1152 ((S)-(+)-2 -Methyl-1-[(4-methyl-5-isoquinolinyl) sulfonyl] -hexahydro-1H-1,4-diazepine) or its dihydrochloride (eg Sasaki et al., Pharmacol. Ther. 93: 225-232) (See 2002), Wf-536 ((+)-(R) -4- (1-aminoethyl) -N- (4-pyridyl) benzamide monohydrochloride) (for example, Nakajima et al., Cancer Chemother. Pharmacol. 52 (4): 319-324 (2003)) and those Derivatives, as well as antisense nucleic acid to ROCK, RNA interference-inducing nucleic acid (e.g., siRNA), dominant negative mutants, and their expression vectors thereof. In addition, since other low molecular weight compounds are known as ROCK inhibitors, such compounds or their derivatives can also be used in the present invention (for example, US Patent Application Publication Nos. 20050209261 and 20050192304, No. 20040014755, No. 20040002508, No. 20040002507, No. 20030125344, No. 20030087919, and International Publication Nos. 2003/062227, 2003-059913, 2003/062225, and 2002/076976, 2004/039796). In the present invention, at least one ROCK inhibitor may be used. ROCK inhibitors are generally used in the range of 100 nM to 50 μM.
 上記の培養細胞の製造方法は,無血清培地が,EGFをさらに含むことが好ましい。無血清培地がEGFを含むと,神経幹細胞が維持され, NestinのmRNAが高発現する。 In the method for producing cultured cells described above, the serum-free medium preferably further comprises EGF. When serum-free medium contains EGF, neural stem cells are maintained and Nestin mRNA is highly expressed.
 EGFは,上皮増殖因子(EGF:epidermal  growth  factor)ファミリーから選択される増殖因子である。例えば,特許6191694号公報では,培地に対するEGFの含有量が,終濃度で,0.5~200ng/mLとされている。 EGF is a growth factor selected from the epidermal growth factor (EGF) family. For example, in Japanese Patent No. 6191694, the content of EGF in the medium is set to 0.5 to 200 ng / mL as the final concentration.
 GFAP
 GFAP遺伝子は,脊損への移植に適しているとされる成体神経幹細胞(adult neural stem cell)または星状細胞(astrocytes)のマーカーである(非特許文献1,20,22)。
GFAP
The GFAP gene is a marker for adult neural stem cells or astrocytes that is considered to be suitable for transplantation into spinal cord injury ( Non-patent Documents 1, 20, 22).
 上記の培養細胞の製造方法は,無血清培地が,FGF2をさらに含むことが好ましい。無血清培地がROCK阻害剤を含むと,実施例において示された通り,EGFのmRNAが高発現し,さらにCD73タンパク質が発現する。FGF(線維芽細胞増殖因子)2は,FGF-2とも表記される。例えば,特許5856029号公報において,FGF2は,0.1~50ng/mL,より好ましくは1~10ng/mL,最も好ましくは5ng/mLの濃度において,培地に添加されることとされている。CD73(エクト-5‘-ヌクレオチダーゼ:Ecto-5’-nucleotidase)は,脊損患者に特有な疼痛を緩和する可能性が高いとされている。 In the method for producing cultured cells described above, the serum-free medium preferably further contains FGF2. When the serum-free medium contains a ROCK inhibitor, EGF mRNA is highly expressed and CD73 protein is further expressed, as shown in the examples. FGF (fibroblast growth factor) 2 is also referred to as FGF-2. For example, in Japanese Patent No. 5856029, FGF2 is added to the medium at a concentration of 0.1 to 50 ng / mL, more preferably 1 to 10 ng / mL, most preferably 5 ng / mL. CD73 (Ecto-5'-nucleotidase: Ecto-5'-nucleotidase) is considered to be highly likely to relieve pain typical of patients with back pain.
 上記の培養細胞の製造方法は,無血清培地が,のカルシウム・イオン濃度が0.03mM以上0.12mM以下であることが好ましい。この範囲に,カルシウム・イオン濃度を維持することで,幹細胞の分化を抑制できる。 In the above-mentioned method for producing cultured cells, the serum-free medium preferably has a calcium ion concentration of not less than 0.03 mM and not more than 0.12 mM. By maintaining the calcium ion concentration within this range, stem cell differentiation can be suppressed.
 なお,上記の好ましいとされた態様は,適宜組み合わせることができる。上記したSAG,パルモルファミン,ソニック・ヘッジホッグ (SHH)タンパク質,B-27サプリメント,ROCK阻害剤,EGF,FGF2などの添加物は,それらの物質の性質を考慮し,適切な量を培地に添加すればよい。これらの物質は,培地に添加されるものとして公知であるから,公知の資料に基づき,添加量を適宜調整すればよい。たとえば,それぞれ培地に0.1ng/mL以上20μg/mL以下(又は0.2ng/mL以上10μg/mL以下)となるように添加すればよい。これらは精製の程度や必要量に応じて適宜調整して添加すればよい。 The above preferred embodiments can be combined as appropriate. Additives such as SAG, palmorphamine, sonic hedgehog (SHH) protein, B-27 supplement, ROCK inhibitor, EGF, FGF2 etc. should be added to the medium in an appropriate amount, taking into consideration the properties of those substances. It may be added. Since these substances are known to be added to the culture medium, the addition amount may be appropriately adjusted based on known materials. For example, it may be added to the culture medium to be 0.1 ng / mL or more and 20 μg / mL or less (or 0.2 ng / mL or more and 10 μg / mL or less). These may be appropriately adjusted and added according to the degree of purification and the required amount.
 上記の培養細胞の製造方法は,ヒト皮膚由来幹細胞が,採取されたヒト皮膚をディスパーゼ処理,トリプシン処理,及びコラゲナーゼ処理の順で処理して得られた細胞であることが好ましい。ディスパーゼ処理,トリプシン処理,及びコラゲナーゼ処理は,いわゆる酵素処理である。酵素処理条件は,生理学的に許容されるpH,例えば約6~8,好ましくは約7.2~7.6に緩衝された等張の塩溶液,例えばトリプシンの場合はPBSやハンクスのバランス塩溶液中で,例えばディスパーゼ,コラゲナーゼの場合は例えばMEM培地中で,約4~40℃,好ましくは約4~39℃で,結合組織を分解するために十分な時間,例えば約1~1000分間,好ましくは5~720分間で,そのために十分な濃度,例えば約0.0001~5%w/v,好ましくは約0.001%~0.5% w/vであり得る。 In the method of producing cultured cells described above, preferably, human skin-derived stem cells are cells obtained by treating collected human skin in the order of dispase treatment, trypsin treatment, and collagenase treatment. Dispase treatment, trypsin treatment, and collagenase treatment are so-called enzyme treatments. The enzyme treatment conditions may be an isotonic salt solution buffered to a physiologically acceptable pH, such as about 6 to 8, preferably about 7.2 to 7.6, such as PBS or Hanks balance salt in the case of trypsin. In solution, eg dispase, collagenase, eg in MEM medium, at about 4-40 ° C., preferably about 4-39 ° C., for a time sufficient to degrade connective tissue, eg about 1-1000 minutes, Preferably, for 5 to 720 minutes, the concentration may be sufficient for that, for example about 0.0001 to 5% w / v, preferably about 0.001% to 0.5% w / v.
 本発明の第2の側面は,脊髄損傷疾患の治療剤の製造方法をも提供する。脊髄は脊椎の中を通る中枢神経であり,この神経を修復するために用いられるのが脊髄損傷疾患の治療剤である。この方法は,上記の方法を用いて培養細胞を製造する。その後,培養細胞,培養工程において得られた培地,及び培養工程において得られた分泌物(エクソソーム等)のいずれか1種又は2種以上を有効成分とした脊髄損傷疾患の治療剤(本発明の剤)を製造する。分泌物(エクソソーム等)は培養上清由来のものであってもよい。また,培地は培養上清のみを用いてもよい。 The second aspect of the present invention also provides a method of producing a therapeutic agent for spinal cord injury. The spinal cord is a central nerve that passes through the spine, and it is a therapeutic agent for spinal cord injury that is used to repair the nerve. This method produces cultured cells using the method described above. Thereafter, a therapeutic agent for a spinal cord injury disease, which comprises one or more of a cultured cell, a culture medium obtained in a culture step, and a secretion (exosome etc.) obtained in a culture step as an active ingredient Agent). The secretion (such as exosome) may be from culture supernatant. Also, as the medium, only the culture supernatant may be used.
 本発明の剤は,当業者に公知の方法で製造すればよい。本発明の剤は,経口用製剤および非経口用製剤として製造することができるが,好ましくは非経口用製剤である。このような非経口用製剤は,液剤(水性液剤,非水性液剤,懸濁性液剤,乳濁性液剤など)としてもよいし,固形剤(粉末充填製剤,凍結乾燥製剤など)としてもよい。また,本発明の剤は,徐放製剤としてもよい。生きた細胞を剤とする場合の剤形としては液剤が好ましく,細胞の部分成分もしくは死細胞全体を剤とする場合の剤形としては,液剤も固形剤も選択できる。 The agent of the present invention may be produced by methods known to those skilled in the art. The agent of the present invention can be manufactured as an oral preparation and a parenteral preparation, but is preferably a parenteral preparation. Such a parenteral preparation may be a solution (aqueous solution, non-aqueous solution, suspension solution, emulsion solution, etc.) or a solid agent (powder filled formulation, freeze-dried preparation, etc.). In addition, the agent of the present invention may be in the form of a sustained release preparation. As a dosage form in the case of using living cells as an agent, a solution is preferable, and as a dosage form in the case of using a partial component of cells or whole dead cells, a solution or a solid agent can be selected.
 本発明の剤の主成分は,培養されたヒト皮膚由来幹細胞,培養工程において得られた培地,及び培養細胞からの分泌物(エクソソーム等)のいずれか1種又は2種以上である。いずれにせよ例えば,細胞の数を指標として,有効量を設計すればよい。最終的な剤には,幹細胞が除かれていてもよい。また分泌物は培養上清に含まれるものであってもよい。1回当たりの投与量と投与回数,投与の頻度は,投与する対象,年齢,症状などによって変化する。一般的には,1日のヒト皮膚由来幹細胞剤の投与量の例は,静脈内注射1投与単位として,1×10細胞以上1×109細胞であり,好ましくは1×106細胞以上5×108細胞であり,より好ましくは1×107細胞以上2×108細胞である。または,体重1kg当たりのヒト皮膚由来幹細胞の1日の投与量の例は,1×10細胞以上1×10細胞であり,好ましくは1×10細胞以上5×10細胞であり,より好ましくは1×10細胞以上2×10細胞である。本発明の剤は,3日に1回の投与で合計8回投与しても良いし,連日投与を一週間継続しても良い。また,様々な頻度の投与期間に組合せにおいての使用もできる。1回で症状の改善が認められた著効例においては,初回のみの投与で終了しても良い。このように,本発明の剤は,疾患や症状の背景によって,多種多様な使用が可能であり,さらには,それぞれの投与における移植細胞数は,上述の範囲において,任意に設定が可能である。 The main components of the agent of the present invention are any one or more of cultured human skin-derived stem cells, a culture medium obtained in the culturing step, and secretions from cultured cells (exosome etc.). In any case, for example, the effective amount may be designed using the number of cells as an index. Stem cells may be removed as the final agent. The secretion may also be contained in the culture supernatant. The dose and frequency of administration and the frequency of administration per dose vary depending on the subject, age, symptoms, etc. In general, an example of a daily dose of human skin-derived stem cell agent is 1 × 10 5 cells or more and 1 × 10 9 cells or more, preferably 1 × 10 6 cells or more, as one administration unit for intravenous injection. 5 × 10 8 cells, more preferably 1 × 10 7 cells or more and 2 × 10 8 cells. Alternatively, an example of a daily dose of human skin-derived stem cells per kg body weight is 1 × 10 4 cells or more and 1 × 10 8 cells, preferably 1 × 10 5 cells or more and 5 × 10 7 cells, More preferably, it is 1 × 10 6 cells or more and 2 × 10 7 cells. The agent of the present invention may be administered once every three days for a total of eight times, or daily administration may be continued for one week. It can also be used in combination for various frequency administration periods. In the case of significant response, where improvement of symptoms was observed at one time, it may be ended with only the first administration. Thus, the agent of the present invention can be used in a wide variety of ways depending on the background of the disease or condition, and further, the number of transplanted cells in each administration can be arbitrarily set within the above-mentioned range .
 本発明は,脊髄損傷の治療剤を製造するための上記した本発明のヒト皮膚由来幹細胞の使用をも提供する。 The present invention also provides the use of the above-mentioned human skin-derived stem cells of the present invention for producing a therapeutic agent for spinal cord injury.
 本発明は脊髄損傷の治療方法をも提供する。各種疾患の治療方法は,脊髄損傷に罹患した患者に上記のように製造された本発明の剤を,有効量投与する工程を含む。 The invention also provides a method of treating spinal cord injury. The method for treating various diseases comprises the step of administering an effective amount of the agent of the present invention produced as described above to a patient suffering from spinal cord injury.
 本発明は,ウィルスによる遺伝子導入法を採用せずに,安全なGDNF遺伝子発現(mRNA)の増加を行う細胞培養方法を提供できる。 The present invention can provide a cell culture method for safely increasing GDNF gene expression (mRNA) without employing a viral gene transfer method.
 本発明は,神経幹細胞のマーカーなど種々のマーカーをも発現する細胞培養方法を提供できる。 The present invention can provide a cell culture method which also expresses various markers such as neural stem cell markers.
 本発明は,脊髄損傷疾患の治療剤の製造方法を提供できる。 The present invention can provide a method for producing a therapeutic agent for spinal cord injury.
 本発明の方法では,ヒトの組織の中でもとりわけ採取しやすい皮膚から脊髄損傷の治療用細胞を無血清・無フィーダー細胞培養法で得ることができる。本発明の方法では,脊髄損傷の治療に最も必須とされるGDNFの発現量を10倍あげる方法を見出し,これと共に,神経幹細胞のマーカーのネスチンを発現させ,脊髄損傷の治療に有効とされるGFAPを発現させ,更に脊髄損傷で起こる痛覚過敏を防ぐCD73も発現させた脊髄損傷治癒に効果の高い細胞を遺伝子導入無しで得ることが確認された。本発明の方法で得られた細胞は,ラット脊髄損傷モデルで治癒効果が確認された。また,この細胞は発癌性試験で陰性であった。更に,この細胞を月2回腰椎穿刺法で投与することにより2年以上脊髄損傷(Th3)で歩けなかった47歳の患者を投与1か月後に歩行可能となる結果を得ることが出来た。一方,2014年のTRIシンポジウムで発表された日本での臭粘膜移植では35歳以下下肢完全麻痺(Th4~7)4例に対し1例の歩行可能例もなかった。 In the method of the present invention, cells for treatment of spinal cord injury can be obtained from the skin which is easy to be collected among human tissues by the serum-free / feeder-free cell culture method. In the method of the present invention, a method of increasing the expression level of GDNF which is most essential for treatment of spinal cord injury by 10 times is found, together with this, nestin of neural stem cell marker is expressed to be effective for treatment of spinal cord injury It was confirmed that cells that are highly effective in healing spinal cord injury, in which CDAP is also expressed, which expresses GFAP and prevents hyperalgesia caused by spinal cord injury, without gene transfer. The cells obtained by the method of the present invention were confirmed to have a healing effect in a rat spinal cord injury model. Also, these cells were negative in oncogenic tests. Furthermore, by administering these cells twice a month by lumbar puncture, it was possible to obtain a result that a 47-year-old patient who could not walk with spinal cord injury (Th3) for more than 2 years could be walked one month after administration. On the other hand, in the odorous membrane transplantation in Japan announced at the 2014 TRI Symposium, there was no one case that could be walked for 4 cases of complete leg paralysis (Th4 ~ 7) under the age of 35 years.
図1は,GDNF mRNAの発現量増加効果 (実験例2)を示す図面に替わるグラフである。FIG. 1 is a graph replacing the drawing showing the effect of increasing the expression level of GDNF mRNA (Experimental Example 2). 図2は,Nestin mRNAの発現 (実験例3) を示す図面に替わるグラフである。FIG. 2 is a graph replaced with a drawing showing expression of Nestin mRNA (Experimental Example 3). 図3は,GFAP mRNAの発現 (実験例4) を示す図面に替わるグラフである。FIG. 3 is a graph replaced with a drawing showing expression of GFAP mRNA (Experimental Example 4). 図4は,CD73の発現(CD発現プロフィル)(実験例5) を示す図面に替わるグラフである。FIG. 4 is a graph replaced with a drawing showing expression of CD73 (CD expression profile) (Experimental Example 5). 図5は,ラット脊髄損傷モデルでの効果(BBB score)(実験例6)を示す図面に替わるグラフである。FIG. 5 is a graph replacing the drawing showing the effect (BBB score) (Experimental Example 6) in a rat spinal cord injury model. 図6は,発癌性の評価を示す図面に替わるグラフである。FIG. 6 is a graph replacing a drawing showing evaluation of carcinogenicity.
 以下,本発明を実施するための形態について説明する。本発明は,以下に説明する形態に限定されるものではなく,以下の形態から当業者が自明な範囲で適宜修正したものも含む。 Hereinafter, modes for carrying out the present invention will be described. The present invention is not limited to the embodiments described below, and includes those appropriately modified by the person skilled in the art from the following embodiments within the obvious scope.
 本発明は,ヒト皮膚から皮膚を採取し,組織を除菌する組織採取工程:
細胞採取播種工程:
細胞増殖工程:
継代工程:
分化誘導工程:
移植・加工工程:
以上の工程で脊髄損傷に効果の高い細胞を得て,直接に投与する細胞を提供する方法または他の基剤と結合させた医療機器のための細胞を提供する方法を提供する。さらに以上の工程で得た培養後培地成分を投与する方法を提供する。
The present invention is a tissue collecting process for collecting skin from human skin and disinfecting the tissue:
Cell collection and seeding process:
Cell proliferation process:
Passage process:
Differentiation induction process:
Implantation / processing process:
The present invention provides a method for obtaining cells that are highly effective for spinal cord injury through the above steps, and for providing cells to be directly administered, or for providing cells for a medical device combined with another base. Furthermore, a method of administering the post-culture medium component obtained in the above steps is provided.
 以下,各工程について,その詳細を例をもって説明する。以下の説明は例であり,本発明は以下の例に限定されるものではなく,当業者にとって自明な範囲で条件を適宜変化させたものも含まれる。なお,uはマイクロを意味する場合がある。 Hereinafter, the details of each process will be described using an example. The following description is an example, and the present invention is not limited to the following examples, and includes those in which conditions are appropriately changed within the obvious range for those skilled in the art. In addition, u may mean micro.
 (組織採取工程)
 本工程で採取される皮膚は首から上の部位が望ましく,更に採取後の美容上の観点からは耳介後部の皮膚が最も望ましい。皮膚の性別や年齢は問われなく,70歳代までは可能である。以下の工程は全て無菌操作で行う。採取組織の除菌は0.1um pore sizeの除菌フィフターで芽胞を取り除いた70%エタノール水にて行う。
(Tissue collection process)
The skin collected in this step is desirably located above the neck, and the skin on the back of the pinna is most preferable from the cosmetic point of view after collection. The sex and age of the skin are not limited, and can be up to the 70s. The following steps are all performed aseptically. Bacterial removal of the collected tissue is carried out with 70% ethanol water from which spores have been removed with a 0.1 um pore size removal filter.
 (細胞採取播種工程)
カットした組織を10uM Y-27632の入りのDispase溶液(PBS(-))に,4℃で1晩浸漬する。この組織を次に10uM Y-27632の入りのTrypsin溶液(PBS(-))に37℃,20分浸漬する。さらに,この組織をコラゲナーゼ溶液(DMEM)に浸漬し,37℃,1~2時間スターラーで撹拌して皮膚を消化する。この撹拌後の懸濁液を100umのメッシュを通して残渣を取り除く。この液をPBS(-)にて3回遠心・洗浄して酵素を除き,細胞を得る。この細胞をTrypsin Inhibitor中に入れ10分静置する。この細胞から遠心してInhibitorを除いた後,10uM Y-27632の入った培地に懸濁し,培養皿に播種する。
(Cell collection and seeding process)
The cut tissue is immersed in 10 μM Y-27632 in Dispase solution (PBS (−)) at 4 ° C. overnight. The tissue is then immersed in Trypsin solution (PBS (−)) containing 10 uM Y-27632 at 37 ° C. for 20 minutes. Furthermore, the tissue is immersed in collagenase solution (DMEM) and stirred at 37 ° C. for 1 to 2 hours with a stirrer to digest the skin. Remove the residue from the stirred suspension through a 100 um mesh. The solution is centrifuged and washed three times with PBS (-) to remove the enzyme and obtain cells. The cells are placed in Trypsin Inhibitor and allowed to stand for 10 minutes. The cells are centrifuged to remove the inhibitor, and then suspended in a medium containing 10 uM Y-27632 and seeded in a culture dish.
 (細胞増殖工程)
播種6日までは,培地交換は行わず,培地添加のみとする。その後の培地交換はconditioned medium中に増殖浮遊する細胞を遠心して回収し,新培地に懸濁した細胞を培養系に戻す培地交換を行う。
(Cell proliferation process)
The medium is not changed until the 6th day of seeding, and only the medium is added. The subsequent medium exchange is carried out by centrifuging and recovering the cells floating in the conditioned medium, and the medium suspended in the new medium is returned to the culture system.
 (継代工程)
細胞を30分以上10uM Y-27632処理を行い,EDTAとTrypLEx Expressで細胞をdishより回収する。次にTrypsin Inhibitor中に入れ10分静置する。細胞から遠心してInhibitorを除いた後,10uM Y-27632の入った培地に懸濁し,元の4~5倍の面積のdishに播種する。継代1代目に5枚の100mm dishに培養し,confluenceに達したら,その3枚を移植に用いるか,移植まで凍結保存する。残り2枚を更に6枚の100mm dishに継代培養し,confluenceに達したら因子添加誘導工程に移る。
(Passage process)
The cells are treated with 10 uM Y-27632 for 30 minutes or more, and the cells are recovered from the dish with EDTA and TrypLEx Express. Then place in Trypsin Inhibitor and let stand for 10 minutes. After centrifuging the cells to remove the Inhibitor, the cells are suspended in a medium containing 10 uM Y-27632 and seeded in the original dish of 4-5 times area. The cells are cultured in five 100 mm 2 dishes at passage 1 and when confluence is reached, three of them are used for transplantation or cryopreserved until transplantation. The remaining 2 sheets are further subcultured to 6 100 mm 2 dishes, and when confluence is reached, the process proceeds to the factor addition induction step.
 (因子添加誘導工程)
因子添加誘導工程は,移植前3日目に必要な細胞に薬物処理を行い必要な因子を添加し培養する工程である。
本工程によって,GDNFmRNAを高発現した細胞,例えば,通常培養の10倍以上発現した細胞が得られる。また,本工程によって,CD13が陰性の細胞(CD13(-))が得られる。また,本工程によって,CD34が陰性の細胞(CD34(-))が得られる。また,本工程によって,CD45が陰性の細胞(CD45(-))が得られる。また,本工程によって,CD90が陰性の細胞が得られる(CD90(-))。また,本工程によって, CD73が陽性の細胞(CD73(+))が得られる。培養細胞のうち,CD73が陽性の細胞(CD73(+))の割合(細胞数)の例は,20%以上100%以下でもよいし,40%以上100%以下でもよいし,60%以上99%以下でもよいし,80%以上99%以下でもよい。因子添加誘導工程を終えた細胞は3日後に移植に用いるか,移植まで凍結保存することが好ましい。
(移植・加工工程)
各行程を経て治療必要数に達した細胞を30分以上10uM Y-27632処理を行い,EDTAとTrypLEx Expressで細胞をdishより回収する。次にTrypsin Inhibitor中に入れ10分静置する。細胞から遠心してInhibitorを除いた後,脊髄穿刺で移植する場合は,10cell/ml濃度で臨床用輸液に懸濁して用いる。この細胞は他の基剤や細胞との加工に供する。
(Factor addition induction process)
The factor addition induction step is a step of carrying out drug treatment on required cells on the third day before transplantation, adding necessary factors, and culturing.
By this step, cells in which GDNF mRNA is highly expressed, for example, cells in which 10 or more times normal culture is expressed are obtained. In addition, this step produces CD13 negative cells (CD13 (−)). In addition, this step produces CD34 negative cells (CD34 (−)). In addition, this step produces CD45 negative cells (CD45 (−)). In addition, cells negative for CD90 can be obtained by this step (CD90 (−)). In addition, CD73-positive cells (CD73 (+)) are obtained by this step. 20% or more and 100% or less, or 40% or more and 100% or less, or 60% or more and 99% or less of the percentage (cell number) of cells (CD 73 (+)) positive in CD73 among the cultured cells % Or less, or 80% or more and 99% or less. It is preferable to use cells for which transplantation has been completed for 3 days after transplantation, or to cryopreserve the cells until transplantation.
(Transplanting and processing process)
The cells that have reached the required number for treatment after each treatment are treated with 10 uM Y-27632 for 30 minutes or more, and the cells are recovered from dish with EDTA and TrypLEx Express. Then place in Trypsin Inhibitor and let stand for 10 minutes. After centrifuging the cells to remove the inhibitor, when transplanting by spinal cord puncture, it is used after suspension in a clinical fluid at a concentration of 10 7 cells / ml. The cells are subjected to processing with other carriers and cells.
 以下,実施例を用いて本発明を具体的に説明する。本発明は,以下の実施例に基づいて当業者が公知の技術を用いて適宜調整できる範囲も含む。 Hereinafter, the present invention will be specifically described using examples. The present invention also includes a range that can be appropriately adjusted by those skilled in the art using known techniques based on the following examples.
 (ヒト皮膚からの皮膚幹細胞の採取・培養)
(組織の採取)
 56歳女性のリフト・アップ美容整形手術時に顔部から切除廃棄された約2cm2の皮膚組織の全層部分を用いた。以下の工程は全て無菌操作で行った。この採取組織を0.1um (ミクロン)ポアサイズ(pore size)の除菌フィフターで除菌された70%エタノール水に30秒間浸漬して,直ちにリン酸緩衝食塩水(Phosphate buffer saline)(PBS(-))にて3回洗浄してエタノールを洗浄した。この組織から毛根が存在する箇所を残して皮下脂肪及び真皮結合織を可及的に鋏で除き,次にメスで1mm2程度の大きさにカットした。
(Collection and culture of skin stem cells from human skin)
(Collection of tissue)
A full-thickness area of about 2 cm 2 of skin tissue resected and discarded from the face during lift-up cosmetic surgery of a 56-year-old woman was used. The following steps were all performed aseptically. This collected tissue is immersed in 70% ethanol water disinfected with a 0.1 um (micron) pore size disinfectant for 30 seconds, and immediately phosphate buffer saline (Phosphate buffer saline) (PBS (-) The ethanol was washed three times with. The subcutaneous fat and the dermal connective tissue were removed as much as possible from this tissue except for the places where hair roots were present, and then cut into a size of about 1 mm 2 with a scalpel.
 (細胞採取播種工程1)
  カットした組織を10uM Y-27632の入りの予備冷却したディスパーゼ(Dispase)溶液(25 units/ml of PBS(-))5mlに,4℃で1晩浸漬した。
(Cell collection seeding step 1)
The cut tissue was immersed in 5 ml of a precooled Dispase solution (25 units / ml of PBS (-)) containing 10 uM Y-27632 overnight at 4 ° C.
 (細胞採取播種工程2)
これを遠心して上清を除いた後に,10uM Y-27632の入りのTrypLE Express溶液(5x希釈PBS(-))に37℃,20分浸漬した。
(Cell collection seeding step 2)
After centrifuging this and removing the supernatant, it was immersed in TrypLE Express solution (5 × diluted PBS (−)) containing 10 uM Y-27632 at 37 ° C. for 20 minutes.
 (細胞採取播種工程3)
この組織を遠心して上清を除いた後に,20mlのcollagenase溶液(GIBCO: type I)(1400 units/ ml DMEM培地)に浸漬し,37℃,1.5時間スターラーで撹拌した。
(Cell collection seeding step 3)
The tissue was centrifuged to remove the supernatant, and then immersed in 20 ml of collagenase solution (GIBCO: type I) (1400 units / ml DMEM medium) and stirred with a stirrer at 37 ° C. for 1.5 hours.
 (細胞採取播種工程4)
撹拌後の懸濁液を100umのメッシュを通して未消化の残渣を取り除いた。この液をPBS(-)にて3回遠心・洗浄して酵素を除き,細胞を得た。
(Cell collection seeding step 4)
The suspension after stirring was passed through a 100 um mesh to remove undigested residues. The solution was centrifuged and washed three times with PBS (-) to remove the enzyme, to obtain cells.
(細胞採取播種工程5)
この細胞を大豆トリプシン阻害剤(Soy bean Trypsin Inhibitor)(0.25%/PBS(-))5ml中に入れ10分静置した。
(Cell collection seeding step 5)
The cells were placed in 5 ml of soybean trypsin inhibitor (Soy bean Trypsin Inhibitor) (0.25% / PBS (-)) and allowed to stand for 10 minutes.
 (細胞採取播種工程6)
細胞から遠心してInhibitorを除いた後,10uM Y-27632添加培養培地に懸濁し,培養皿に播種した。出発組織の広さ(1~2 cm)から,1枚の6 well plate (CellBind :COSTAR)の全wellに播種した。
(Cell collection seeding process 6)
The cells were centrifuged to remove the inhibitor, and then suspended in 10 uM Y-27632-added culture medium and seeded in culture dishes. From the size of the starting tissue (1 to 2 cm 2 ), it was seeded to all wells of one 6 well plate (CellBind: COSTAR).
 (培地調製工程)
 培養培地は以下のとおり調製された。すなわち,基本培地のMCDB153培地(Sigma:Stemline Keratinocyte Basal Medium)中に,インスリン(10mg/L), ホロ・トランスフェリン (5.5mg/L), セレニウム(6.7ug/L),エタノールアミン(2mg/L), ビタミンC(L-Ascorbic acid 2-phosphate semimagnesium salt) (50ug/L), KGF (10ug/L), 脂質(脂肪酸混合物:アラキドン酸(20ug/L),コレステロール(2.2mg/L), DL-a-Tocopherol-acetate (700ug/L), リノール酸(100ug/L),リノレン酸(100ug/L),ミリスチン酸(100ug/L),オレイン酸(100ug/L),パルミトレイン酸(100ug/L),パルミチン酸(100ug/L),ステアリン酸(100ug/L),Tween80(22mg/L), Pluronic F-68(1000mg/L), EGF(20ug/L), FGF(10ug/L),Y-27632(1uM), 1%B-27 supplement XenoFree CTS (Invitrogen)を添加して調製した。
(Media preparation process)
Culture medium was prepared as follows. Specifically, insulin (10 mg / L), holo-transferrin (5.5 mg / L), selenium (6.7 ug / L), ethanolamine (2 mg / L) in MCDB 153 medium (Sigma: Stemline Keratinocyte Basal Medium) as a basic medium. , Vitamin C (L-Ascorbic acid 2-phosphate semimagnesium salt) (50 ug / L), KGF (10 ug / L), lipid (a mixture of fatty acid: arachidonic acid (20 ug / L), cholesterol (2.2 mg / L), DL- a-Tocopherol-acetate (700 ug / L), linoleic acid (100 ug / L), linolenic acid (100 ug / L), myristic acid (100 ug / L), oleic acid (100 ug / L), palmitoleic acid (100 ug / L) , Palmitic acid (100 ug / L), stearic acid (100 ug / L), Tween 80 (22 mg / L), Pluronic F-68 (1000 mg / L), EGF (20 ug / L), FGF (10 ug / L), Y- Prepared by adding 27632 (1 uM), 1% B-27 supplement XenoFree CTS (Invitrogen).
 (細胞増殖工程1)
 初代培養は,培養7日までは培地交換をすることなく1/3量の割合で培地添加しつつ馴化培養を行った。その後は2日毎に培地交換しながら培養を行った。この培地交換では,古い培地を遠心し,その2/3量を捨て,残った1/3量の培地と遠心で沈殿した浮遊細胞を混ぜて2/3量の新しい培地をさらに加えて培養皿に戻した。100%コンフルエンスに達するまでに10日間を要した。
(Cell proliferation step 1)
In primary culture, conditioned culture was performed while adding medium at a rate of 1/3 volume without changing the medium until 7 days of culture. After that, culture was performed while changing the medium every two days. In this medium exchange, the old medium is centrifuged, 2/3 of its volume is discarded, the remaining 1/3 volume of medium and the floating cells precipitated by centrifugation are mixed, and 2/3 volume of new medium is further added to the culture dish. I'm back to. It took 10 days to reach 100% confluence.
 (継代工程)
 培養された細胞の培地にY-27632 を10uM濃度で入れて,30分,37℃で静置した。次に培地を全て除き,0.05%EDTA/PBS(-)を2ml/well加えてを10分,37℃で静置した。さらにPBS(-)にて5倍希釈したTrypLEx Expressを0.5ml/well加えてを5分,37℃で静置した。次にピペッティングで細胞をwellより剥離した。ここに2.5ml/wellのTrypsin Inhibitor(0.05%/PBS(-))を加えて回収された細胞を遠心した。上清を除き,2.5mlのTrypsin Inhibitor(0.25%/PBS(-))を入れて細胞を懸濁し10分静置した。細胞から遠心してInhibitorを除いた後,10uM Y-27632の入った培地に懸濁し,培養皿に元の5倍の面積の6 well plate (CellBind :COSTAR)5枚に播種した。1週間後に100%コンフルエンスに達したので,以下の実施例の実験の材料とした。
(Passage process)
Y-27632 was added at 10 uM concentration to the culture medium of the cultured cells and allowed to stand at 37 ° C. for 30 minutes. Next, the medium was completely removed, and 2 ml / well of 0.05% EDTA / PBS (-) was added and allowed to stand at 37 ° C. for 10 minutes. Furthermore, 0.5 ml / well of TrypLEx Express diluted 5-fold with PBS (−) was added, and the mixture was allowed to stand at 37 ° C. for 5 minutes. Next, the cells were detached from the wells by pipetting. To this, 2.5 ml / well of Trypsin Inhibitor (0.05% / PBS (-)) was added, and the recovered cells were centrifuged. The supernatant was removed, 2.5 ml of Trypsin Inhibitor (0.25% / PBS (-)) was added to suspend the cells, and allowed to stand for 10 minutes. After centrifuging the cells to remove the inhibitor, the cells were suspended in a medium containing 10 uM Y-27632, and seeded in culture dishes on 5 pieces of 6-well plate (CellBind: COSTAR) having a 5-fold area of the original. One week after reaching 100% confluence, it was used as the material of the experiment of the following example.
 (GDNF (Glial cell-derived neurotrophic factor) mRNAの発現量増加法)
 細胞のGDNF (Glial cell-derived neurotrophic factor) mRNA発現を増加させる化合物またはサイトカインを見出すために可能性のある以下の6個のものを検討した。Valproic acid (VA) , Platelete-derived growth factor (PDGF), SAG(又はPurmorphamine or rh-Sonic Hedgehog (PeproTech)), Bone morphogenic protein 4 (BMP4) これら6個のものを以下の組合せで実施例1コンフルエントになった7wellに添加した。(1)Control(無添加)(2)VA(1uM) (3)VA(1uM) + PDGF(10ng/ml) (4)VA(1uM)+SAG(0.3uM)  (5)PDGF(10ng/ml) (6) SAG(0.3uM) (7)MBP-4(10ng/ml):添加後72時間後に各wellからTRIzol法にてtotal RNAを抽出した。PCR用cDNA合成キット「ReverTra Ace(登録商標)qPCR RT Kit」を用いて,ReverTra Ace(登録商標) qPCR RT Kitの10μl反応系に対して,各 total RNAを0.5ug でそれぞれ添加し,各cDNA合成を行った。
これらのcDNAサンプルでのGDNF mRNAの発現比較をヒトGDNF リアルタイムPCRプライマーセット(Roche 5326257)を用いてリアルタイムPCR法にてアプラ イドバイオシステムズ社のABI PRISM(登録商標)7700を用いたインターカレーターアッセイ法で行った。qRT-PCR反応はTOYOBOのSYBR(登録商標) Green Realtime PCR Master Mix -Plus- を用いて反応液を調製した。  
反応液の調製  
蒸留水                            11 μL 
SYBR(登録商標) Green Realtime PCR Master Mix -Plus-      25 μL 
Plus solution                         5μL 
プライマー 1 (10μM)                     2 μL 
プライマー 2 (10μM)                     2 μL 
サンプル溶液                                       5 μL 
合計液量                                          50 μL
反応液を96 well plateの各wellに入れてPCR反応を行った。
PCRサイクルは3ステップ法で,アニール 60℃/伸長 72℃を条件とし(1)~(2)を40 cycle行った。最後のステップで融解曲線(Melting Curve)解析を行い,primer dimerの形成が見られないことを確認した。
 95℃ 60秒  
(1)↓
(2)95℃ 15秒  
(3)60℃ 15秒 
(4)72℃ 45秒(data collection)  
また,同時にGAPDHmRNAの検出を行い,GDNFmRNAの発現量を,GDNFmRNA/ GAPDHmRNAとして補正した。その結果,(6)SAGのみが(1)コントロールの10倍以上のGDNFmRNA発現が得られた。(6)のSAGの代わりにPurmorphamine(1.5uM)や, rh-Sonic Hedgehog(10ng/ml)でも同様に10倍近くのGDNFmRNA発現が得られた。(図-1)GAPDHのprimer 配列は以下の配列で合成した。
Forward primer:ATCTTCTTTTGCGTCGCC  (配列番号1)
Reverse primer:GATGACAAGCTTCCCGTTC (配列番号2)
発現鎖長:250bp
(Method to increase the expression level of Glial cell-derived neurotrophic factor (GDNF))
The following six possibilities were examined to find compounds or cytokines that increase cellular GLDF (Glial cell-derived neurotrophic factor) mRNA expression. Valproic acid (VA), Platelete-derived growth factor (PDGF), SAG (or Purmorphamine or rh-Sonic Hedgehog (PeproTech)), Bone morphogenic protein 4 (BMP 4) Example 1 in the following combination of these six: The solution was added to the 7 wells. (1) Control (no additive) (2) VA (1 uM) (3) VA (1 uM) + PDGF (10 ng / ml) (4) VA (1 uM) + SAG (0.3 uM) (5) PDGF (10 ng / ml) (6) SAG (0.3 uM) (7) MBP-4 (10 ng / ml): 72 hours after addition, total RNA was extracted from each well by the TRIzol method. Add 0.5ug of each total RNA to the 10μl reaction system of ReverTra Ace qPCR RT Kit using the cDNA synthesis kit for PCR "ReverTra Ace qq RT Kit", and add each cDNA The synthesis was done.
Comparison of GDNF mRNA expression in these cDNA samples by real-time PCR using human GDNF real-time PCR primer set (Roche 5326257) Intercalator assay using ABI PRISM® 7700 from Aply Biosystems I went there. The qRT-PCR reaction was prepared using TOYOBO's SYBR® Green Realtime PCR Master Mix-Plus-.
Preparation of reaction solution
Distilled water 11 μL
SYBR® Green Realtime PCR Master Mix -Plus- 25 μL
Plus solution 5μL
Primer 1 (10 μM) 2 μL
Primer 2 (10 μM) 2 μL
Sample solution 5 μL
Total volume 50 μL
The reaction solution was placed in each well of a 96 well plate to carry out a PCR reaction.
The PCR cycle was a 3-step method, and 40 cycles of (1) to (2) were performed under the conditions of annealing 60 ° C / extension 72 ° C. Melting curve analysis was performed at the last step to confirm that no formation of primer dimer was observed.
95 ° C 60 seconds
(1) ↓
(2) 95 ° C 15 seconds
(3) 60 ° C 15 seconds
(4) 72 ° C 45 seconds (data collection)
At the same time, GAPDH mRNA was detected, and the expression level of GDNF mRNA was corrected as GDNF mRNA / GAPDH mRNA. As a result, (6) only SAG (1) GDNF mRNA expression more than 10 times of control was obtained. In the case of Purmorphamine (1.5 uM) or rh-Sonic Hedgehog (10 ng / ml) instead of SAG of (6), GDNF mRNA expression near 10 times was obtained similarly. (FIG. 1) The primer sequence of GAPDH was synthesized with the following sequence.
Forward primer: ATCTTCTTTTGCGTGCC (SEQ ID NO: 1)
Reverse primer: GATGACAAAGCTTCCCGTTC (SEQ ID NO: 2)
Expression chain length: 250 bp
 図1は,実施例2におけるGDNF mRNAの発現量の測定結果を示す図面に替わるグラフである。 FIG. 1 is a graph replaced with a drawing showing the measurement results of the expression level of GDNF mRNA in Example 2.
 (Nestin mRNAの神経分化誘導による発現変化)
 実施例1でコンフルエンスに達した3wellsのうち2 wellsに神経前駆細胞の分化同定用キット Human/Mouse/Rat Neural Progenitor Cell Functional Identification Kit(R&D Systems SC082)の分化用培地(medium for NPC differentiation)で7日間培養した。もう一つのwellには発明の維持培地で同日間培養した。7日目に実施例2と同様にtotal RNAを抽出後,cDNAを調製した。このcDNAを用いて神経幹細胞のマーカーであるNestin mRNAの発現比較をRT-PCRにてGAPDH mRNAを基底状態として測定した。Nestin のPrimerはForward CGTTGGAACAGAGGTTGGAG(配列番号3)とReverse TCCTGAAAGCTGAGGGAAG(配列番号4)であり,発現鎖長は262bpであった。GAPDHのprimerは実施例2と同様のものを用いた。
その結果を図2に示す。
(Expression change of Nestin mRNA by induction of neural differentiation)
Kit for differentiation and identification of neural progenitor cells in 2 wells out of 3 wells that reached confluence in Example 1 7 using the culture medium (medium for NPC differentiation) of Neural Progenitor Cell Functional Identification Kit (R & D Systems SC082) It was cultured for a day. The other well was cultured in the maintenance medium of the invention for the same day. After extracting total RNA similarly to Example 2 on the 7th day, cDNA was prepared. Using this cDNA, the expression comparison of Nestin mRNA, which is a marker for neural stem cells, was measured by RT-PCR with GAPDH mRNA as the ground state. The Primer of Nestin was Forward CGTTGGACAAGAGGTTGGAG (SEQ ID NO: 3) and Reverse TCCTGAAAGCTGAGGGAAG (SEQ ID NO: 4), and the expression chain length was 262 bp. The same GAPDH primer as in Example 2 was used.
The results are shown in FIG.
 図2は,実施例3におけるNestin mRNAの発現量を示す図面に替わるグラフである。図2に示されるように,分化培地に変えるとNestinの発現量(Nestin mRNA/GAPDH mRNA)は分化前の1/7程度に減少した。この結果は発明者の増殖培地は神経幹細胞を維持させ,分化培地は神経幹細胞を減少させることを示した。 FIG. 2 is a graph replaced with a drawing showing the expression level of Nestin mRNA in Example 3. As shown in FIG. 2, when changed to the differentiation medium, the expression level of Nestin (Nestin mRNA / GAPDH mRNA) decreased to about 1/7 before differentiation. The results showed that our growth medium maintained neural stem cells, and differentiation medium decreased neural stem cells.
(GFAP mRNAの神経分化誘導による発現変化)
 実施例1でコンフルエンスに達した3wellsのうち2 wellsに神経前駆細胞の分化同定用キット Human/Mouse/Rat Neural Progenitor Cell Functional Identification Kit(R&D Systems SC082)の分化用培地(medium for NPC differentiation)で7日間培養した。もう一のwellには発明の維持培地で同日間培養した。7日目に実施例2と同様にtotal RNAを抽出後,cDNAを調製した。このcDNAを用いてAstrocytes又はAdult neural stem cellsのマーカーであるGFAP mRNAの発現比較をRT-PCRにてGAPDH mRNAを基底状態として測定した。GFAP のPrimerはForward :ACATCGAGATCGCCACCTAC(配列番号5)とReverse :ACATCACATCCTTGTGCTCC(配列番号6)であり,発現鎖長は219bpであった。GAPDHのprimerは実施例2と同様のものを用いた。その結果を図3に示す。
(Expression change of GFAP mRNA by induction of neural differentiation)
Kit for differentiation and identification of neural progenitor cells in 2 wells out of 3 wells that reached confluence in Example 1 7 using the culture medium (medium for NPC differentiation) of Neural Progenitor Cell Functional Identification Kit (R & D Systems SC082) It was cultured for a day. The other well was cultured in the maintenance medium of the invention for the same day. After extracting total RNA similarly to Example 2 on the 7th day, cDNA was prepared. Using this cDNA, the expression comparison of GFAP mRNA, which is a marker of Astrocytes or Adult neural stem cells, was measured by RT-PCR with GAPDH mRNA as the basal state. The GFAP Primer was Forward: ACATCGAGATCGCCCACCTAC (SEQ ID NO: 5) and Reverse: ACATCACATCCTTGTGCTCC (SEQ ID NO: 6), and the expression chain length was 219 bp. The same GAPDH primer as in Example 2 was used. The results are shown in FIG.
 図3は,実施例4におけるGFAP mRNAの発現量を示す図面に替わるグラフである。図4に示されるように,分化培地に変えるとGFAPの発現量(GFAP mRNA/GAPDH mRNA)は分化前より僅かに増加した。この結果は発明者の増殖培地は神経幹細胞を維持させ,その中のGFAPの発現も維持させることを示した。 FIG. 3 is a graph replaced with a drawing showing the expression level of GFAP mRNA in Example 4. As shown in FIG. 4, the expression level of GFAP (GFAP mRNA / GAPDH mRNA) slightly increased when it was changed to the differentiation medium, compared to before differentiation. The results show that the inventors' growth media maintain neural stem cells and also maintain the expression of GFAP in them.
 (フローサイトメトリーによるCD抗原発現検討)
 培養されてコンフルエントになった細胞を培養皿からtrypsin-EDTA溶液を用いて剥離・単離した。次に3%BSA/PBS(-)溶液で30分間置くことによりブロッキングを行い,非特異的吸着を防いだ。更に4%パラホルムアルデヒドで10分固定後,0.5% Triton X-100/PBS(-)に5分,4℃インキュベートした。この細胞をCD73に対するモノクローナル抗体CD73-PE (BD 561014)にて4℃オーバーナイトでインキュベートした。この細胞を5回洗浄液で洗浄して結合しなかった抗体を除去し,フローサイトメトリーはSONY EC800を用いて抗原の発現を解析した。その結果を図4に示す。
(CD antigen expression examination by flow cytometry)
The cultured and confluent cells were detached and isolated from culture dishes using trypsin-EDTA solution. Blocking was then performed by placing in a 3% BSA / PBS (-) solution for 30 minutes to prevent nonspecific adsorption. After further fixing for 10 minutes with 4% paraformaldehyde, the cells were incubated at 4 ° C. for 5 minutes in 0.5% Triton X-100 / PBS (-). The cells were incubated at 4 ° C. overnight with the monoclonal antibody CD73-PE (BD 561014) against CD73. The cells were washed 5 times with a washing solution to remove non-bound antibody, and flow cytometry was performed to analyze the expression of the antigen using SONY EC800. The results are shown in FIG.
 図4は,実施例5における CD73の発現(CD発現プロフィル)を示す図面に替わるグラフである。図4に示されるように,CD13,CD34, CD45がネガティブなことから,この細胞は非血液系細胞であることが示された。また,CD90もネガティブなことから,ヒト皮膚由来間葉系幹細胞でないことも示された。また,CD73が強く発現していることが,この細胞の最もはっきりしたCDの特徴であった。 FIG. 4 is a graph replaced with a drawing showing expression of CD73 (CD expression profile) in Example 5. As shown in FIG. 4, CD13, CD34 and CD45 were negative, indicating that the cells were non-hematologic cells. In addition, CD90 was also negative, indicating that it is not human skin-derived mesenchymal stem cells. In addition, the strong expression of CD73 was the most distinctive feature of CD in this cell.
 (ラット脊髄損傷モデルでのBBB score法による効果検討)
 SAG処理等でGDNFを増加させていない培養細胞を酵素Accutase(GIBCO A11105)を用いて培養皿から剥離した。この剥離細胞1x10個を200ulの培養液に懸濁し,1週間前に脊髄に錘で損傷を与え,かつ免疫抑制剤を投与したラットの脊髄損傷部位に注入移植した。対照のラットには同量の培養培地のみを注入するsham operationを行った。細胞移植群は5匹,対照群は4匹のラットを実験に用いた。脊髄損傷の治療効果は動物の動きをビデオ装置で観察して点数化するBBB Score法で測定した。結果を図に示す。図中,横軸は細胞移植後の日数,縦軸はBBB Scoreを示す。実線は上記細胞を移植した結果,点線は対照の結果を示し,*はStudent T testによる有意差検定の結果P<0.05, **はP<0.01, ***はP<0.001であることを示す。投与後3日から対照群に比較して細胞移植群はBBB Scoreで2点差の付く統計的に有意な効果(P<0.05)を示した。移植後35日を過ぎると,対照群に比較して細胞移植群はBBB Scoreで4点差の付く統計的に有意な効果(P<0.001)を示した。骨髄間質細胞移植の報告(非特許文献23)では,有意差がつくまで2週間近くかかっている。また,iPSから誘導した神経幹細胞移植(非特許文献1)では有意差がつくまで3週間を要している。これらに比べ今回の結果は劇的に早く,3日からの効果が見られた。しかも骨髄間質細胞移植では4週間経過しても,有意差はP<0.05であるが,本発明は同時期にP<0.001とより優位であった。この結果は本発明で得られる細胞が脊髄損傷の治療に非常に効果的であることを示している。
(Investigation of effect by BBB score method in rat spinal cord injury model)
Cultured cells in which GDNF was not increased by SAG treatment or the like were detached from the culture dish using the enzyme Accutase (GIBCO A11105). The exfoliated cells (1 × 10 5 ) were suspended in 200 ul of culture solution, and one week ago, the spinal cord was injured with a weight and injected and implanted at the site of spinal cord injury in a rat treated with an immunosuppressant. Control rats underwent a sham operation in which only the same amount of culture medium was injected. Five rats were in the cell transplantation group, and four rats in the control group were used for the experiment. The therapeutic effect of spinal cord injury was measured by the BBB Score method in which the movement of the animal was observed with a video device and scored. The results are shown in the figure. In the figure, the horizontal axis shows the number of days after cell transplantation, and the vertical axis shows BBB Score. The solid line shows the result of transplantation of the above cells, the dotted line shows the result of the control, * shows that the result of the significant difference test by Student T test is P <0.05, ** is P <0.01 and *** is P <0.001. Show. From the 3rd day after administration, the cell transplantation group showed a statistically significant effect (P <0.05) with a 2-point difference in BBB Score compared to the control group. After 35 days after transplantation, the cell transplantation group showed a statistically significant effect (P <0.001) with a 4-point difference in BBB Score compared to the control group. In the report of bone marrow stromal cell transplantation (Non-patent Document 23), it takes nearly two weeks until a significant difference is obtained. In addition, it takes three weeks for iPS-derived neural stem cell transplantation (Non-patent Document 1) to produce a significant difference. This result is dramatically faster than these, and the effect from 3 days was seen. Moreover, in the bone marrow stromal cell transplantation, even after 4 weeks, the significant difference was P <0.05, but the present invention was more dominant with P <0.001 at the same time. This result shows that the cells obtained by the present invention are very effective in the treatment of spinal cord injury.
 (造腫瘍性の評価)
動物及び試験法
 実験にはNOD/Shi-scid, IL-2Rγnull(登録商標)系統のマウス(6週齢,♀,公共財団法人 実験動物中央研究所)を用いた。1週間の順化期間の後,汎用群分けシステム(株式会社ヴィジョンズ)を用いて,可能な限り体重の平均値が同じくなるようにして3群に群分けした. それぞれの群のマウスの右側腹部に,実施例1と同じ方法で培養した(細胞Aとする),陰性対照細胞として培養液,陽性対照細胞としてのHeLa S3(DSファーマバイオメディカル株式会社)をそれぞれシリンジ(マイジェクター(登録商標),針仕様:27G)により移植(いずれも0.2 mL/個体)した。それぞれの群を細胞A群(10個体),培養液群(10個体),HeLa S3群(5個体)とする。なお,細胞Aは,実施例1と同じ方法で培養した細胞を3系統用意し,α-MEM培地にそれぞれ1x10 cells/mLで懸濁し,空気を含まないように2 mlのクライオチューブに分注した。分注のおよそ3時間後,α-MEM培地を除いて培養培地液に再懸濁し,3系統を等量の細胞数ずつ混合して5x10 cells/mLの懸濁液を調製し,移植まで4℃にて保存し,調製開始後2時間以内に移植を行った。なお,直前に計測した3系統を混合した検体の移植後の生存率は75%であった。
それぞれの群のマウスは,「公益財団法人実験動物中央研究所・本所における実験動物の飼育管理に関する作業基準」に準拠して移植から13週飼育し,一般状態観察,体重測定および結節の観察・サイズ測定を週1回行った。結節を観察する際は,被験物質移植部位を指で触診し,硬い感触のある場合に結節の形成が確認されたものとし,長径(L)と短径(W)をノギスで測定した。結節体積(V)は「ヌードマウスと抗癌剤評価」(野口達次,櫻井欽夫,稲葉實編著,蟹書房,1991年6月)の腫瘍体積簡易計算式を用い,計算式V=LW/2で算出した(単位は長径と短径はmm, 体積はmm). なお,HeLaS3群の個体は,観察期間中に結節重量(比重を1として体積より計算)が体重の1/10を超えたことが確認されたため,第5週~第8週の時点で人道的エンドポイントとして観察を終了し安楽死処分した。
 安楽死処分の際,マウスはイソフルラン麻酔下で全放血により安楽死させ,胸腔内,腹
腔内諸臓器を観察した。また移植部位皮膚を皮下組織を含めて採材し,10%中性緩衝ホル
マリン液で固定した。ホルマリン固定標本は常法にてパラフィン包埋後薄切しHE染色,抗HLA(Human Leucocyte Antigen)免疫染色を行い,光学顕微鏡で観察した。
(Evaluation of tumorigenicity)
Animals and Test Methods For the experiments, mice (6 weeks old, rabbit, Central Research Institute for Experimental Animals) having NOD / Shi-scid, IL-2Rγ null (registered trademark) strain were used. After one week of acclimatization period, using a universal grouping system (Visions Co., Ltd.), groups were divided into three groups with the same average weight value as possible, with the right flank of each group of mice. In the same manner as in Example 1 (cultured as cell A), a culture solution as negative control cells, and HeLa S3 (DS Pharma Biomedical Co., Ltd.) as positive control cells were each syringe (Myjector (registered trademark), Needle specifications (27 G) were transplanted (both in 0.2 mL / individual). Let each group be cell A group (10 individuals), culture fluid group (10 individuals), and HeLa S3 group (5 individuals). For cell A, prepare 3 lines of cells cultured in the same manner as in Example 1, suspend each at 1 × 10 7 cells / mL in α-MEM medium, and divide into 2 ml cryotubes so as not to contain air. I ordered. Approximately 3 hours after pipetting, remove the α-MEM medium and resuspend in the culture medium, mix 3 lines in equal numbers of cells to prepare a suspension of 5 × 10 7 cells / mL, and complete transplantation It was stored at 4 ° C., and transplantation was performed within 2 hours after the start of preparation. The survival rate after transplantation of the sample prepared by mixing the three lines measured immediately before was 75%.
The mice in each group are bred 13 weeks after transplantation in accordance with the “Working standard for breeding and management of experimental animals at the Central Research Institute for Experimental Animals of the Public Interest Foundation”, general condition observation, weight measurement and observation of nodules -Size measurement was performed once a week. When observing a nodule, the test substance transplantation site was palpated with a finger, and the formation of a nodule was confirmed when there was a firm feel, and the major diameter (L) and the minor diameter (W) were measured with a caliper. Nodule volume (V) is using the tumor volume simple formula of "nude mice and the anti-cancer agent evaluation" (Noguchi Itarutsugi, Sakurai欽夫, Minoru Inaba written and edited, crab Shobo, June 1991), the calculation formula V = LW 2 / Calculated in 2 (in units of mm and mm in diameter, mm 3 in volume). Note that individuals in the HeLaS3 group have 1/10 weight of the body weight (the specific gravity is 1 and the volume is calculated from the volume) during the observation period. Since it was confirmed that it was exceeded, observation was ended as a humanitarian endpoint at weeks 5 to 8, and euthanasia was given.
At the time of euthanasia, the mice were euthanized by total bleeding under isoflurane anesthesia, and the chest and abdominal organs were observed. In addition, the skin at the transplantation site was collected including subcutaneous tissue and fixed with 10% neutral buffered formalin solution. Formalin-fixed specimens were paraffin-embedded, sliced, HE staining, anti-HLA (Human Leucocyte Antigen) immunostaining, and observed with a light microscope in a conventional manner.
 結果
 その結果を図6に示す。図6は,発癌性の評価を示す図面に替わるグラフである。HeLa S3群においては,移植後3週で100%(5/5例)の個体に結節の形成が認められたが,培養液群及び細胞A群では観察終了時まで結節の形成は認められなかった。HeLa S3群の全例で結節が形成された。この結節は連続した体積増加を示し低分化型癌の組織像と共に抗HLA反応を示したため,HeLa S3細胞に由来する腫瘍であると判断された。細胞A群では病理組織検査により6/10例に抗HLA反応陽性を示す繊維増殖が確認され,移植細胞が残存したものと判断された。しかし,全観察期間を通じて全例で結節の形成は認められず,病理組織検査におけるHE染色像において過形成変化あるいは腫瘍性変化を認める所見は認められなかった。この結果は,本試験の条件下では細胞Aは造腫瘍性を示さないことを示している。
Results The results are shown in FIG. FIG. 6 is a graph replacing a drawing showing evaluation of carcinogenicity. In the HeLa S3 group, nodule formation was observed in 100% (5 of 5) individuals at 3 weeks after transplantation, but no nodule formation was observed in the culture fluid group and cell group A until the end of observation The Nodules were formed in all cases in the HeLa S3 group. The nodule was considered to be a tumor derived from HeLa S3 cells, as it showed continuous volume increase and showed anti-HLA reaction together with the histological image of poorly differentiated carcinoma. In the cell group A, histopathological examination confirmed fiber proliferation showing anti-HLA positive in 6/10 cases, and it was judged that the transplanted cells remained. However, no nodule formation was observed in all cases throughout the entire observation period, and no finding of hyperplastic or neoplastic changes was found in the HE staining image in histopathological examination. This result indicates that cell A does not exhibit tumorigenicity under the conditions of this test.
 (ヒト脊髄損傷患者での効果検討:GDNFの効果)
 自己細胞を各自己皮膚より培養し,SAG等を用いたGDNFを増加させない培養細胞を酵素Accutase(GIBCO A11105)を用いて培養皿から剥離した。事前に腰椎穿刺で1 mlの脊髄液を採取した後に,1 mlの注射用生理食塩液に懸濁した剥離細胞1x10個を穿刺跡に自家移植した。この症例1~7の脊損患者7名は年齢は32~51歳で,男性6名,女性1名であった。投与回数は2~3回で,投与間隔は1~3か月であった。損傷から投与までの期間は3~23年,C4~7の頚髄損傷で,四肢麻痺であった。副作用は投与日の頭痛・発熱が見られた。7名中3名には治療効果はなかった。他の4名は治療効果が認められ,腕の発汗,皮膚感覚回復,尿の貯留改善,握力アップ,指の動き改善等の自覚症状があった。症例8のみ細胞のSAG処理を行い,GDNF mRNAを増加させた細胞を移植した。上記と同じ細胞数を1か月以内に2回投与した。症例8の患者は47歳男性で,胸髄Th3損傷で,損傷から投与までの期間は2年半,下肢麻痺であった。副作用は投与時の頭痛があった。治療効果は投与1か月後に現れ,補助下での立位歩行が可能となった。症例8は症例1~7に比較して下肢麻痺と軽症で投与までの2年半と短かったが効果は劇的でまた回復時間も短かった。
(Evaluation of effects in patients with human spinal cord injury: Effects of GDNF)
Autologous cells were cultured from each autologous skin, and cultured cells that did not increase GDNF using SAG or the like were detached from the culture dishes using the enzyme Accutase (GIBCO A11105). After 1 ml of spinal fluid was collected by lumbar puncture in advance, 1 × 10 7 detached cells suspended in 1 ml of saline for injection were autologously transplanted into the puncture mark. Seven patients with SCI of this case 1 to 7 were 32 to 51 years old, 6 males and 1 female. The administration frequency was 2 to 3 times, and the administration interval was 1 to 3 months. The period from injury to administration was 3 to 23 years, C4 to 7 cervical cord injury and quadriplegia. The side effects were headache and fever on the day of administration. Three out of seven had no therapeutic effect. The other 4 had therapeutic effects, and had subjective symptoms such as arm sweating, cutaneous sensation recovery, urine retention improvement, grip strength improvement, finger movement improvement, etc. Only in case 8 cells were treated with SAG, and cells with increased GDNF mRNA were transplanted. The same number of cells as above was administered twice within one month. The patient of Case 8 was a 47-year-old man with Th3 injury, and the period from injury to administration was 2.5 years, with leg paralysis. Side effects were headaches at the time of administration. The treatment effect appeared one month after administration, and it became possible to stand on foot under assistance. Case 8 was short and two and a half years before administration with leg paralysis and mildness compared to cases 1 to 7, but the effect was dramatic and recovery time was also short.
 本発明は医薬産業において利用されうる。 The present invention can be utilized in the pharmaceutical industry.
配列番号1:プライマー
配列番号2:プライマー
配列番号3:プライマー
配列番号4:プライマー
配列番号5:プライマー
配列番号6:プライマー
Sequence number 1: Primer sequence number 2: Primer sequence number 3: Primer sequence number 4: Primer sequence number 5: Primer sequence number 6: Primer

Claims (8)

  1.  ヒト皮膚由来幹細胞を,SAGを含む無血清培地で培養する培養工程を含む,グリア細胞株由来神経栄養因子(GDNF)mRNA及びCD73タンパク質を含む培養細胞の製造方法。 A method for producing a cultured cell comprising glial cell line-derived neurotrophic factor (GDNF) mRNA and a CD73 protein, comprising a culture step of culturing human skin-derived stem cells in a serum-free medium containing SAG.
  2.  請求項1に記載の方法であって,
     前記無血清培地は,パルモルファミン(Purmorphamine)及びソニック・ヘッジホッグ (Sonic hedgehog, SHH)タンパク質のいずれか又は両方をさらに含む,方法。
    The method according to claim 1, wherein
    The serum-free medium further comprises either or both of a Parmorphamine (Purmorphamine) and a Sonic hedgehog (SHH) protein.
  3.  請求項1に記載の方法であって,
     前記無血清培地は,B-27サプリメントをさらに含む,方法。
    The method according to claim 1, wherein
    The serum-free medium further comprises a B-27 supplement.
  4.  請求項1に記載の方法であって,
     前記無血清培地は,ROCK阻害剤をさらに含む,方法。
    The method according to claim 1, wherein
    The method wherein the serum-free medium further comprises a ROCK inhibitor.
  5.  請求項1に記載の方法であって,
     前記無血清培地は,EGF又はFGF2をさらに含む,方法。
    The method according to claim 1, wherein
    The method wherein the serum-free medium further comprises EGF or FGF2.
  6.  請求項1に記載の方法であって,
     前記無血清培地は,カルシウム・イオン濃度が0.03mM以上0.12mM以下である,方法。
    The method according to claim 1, wherein
    The serum-free medium has a calcium ion concentration of not less than 0.03 mM and not more than 0.12 mM.
  7.  請求項1に記載の方法であって,
     前記ヒト皮膚由来幹細胞は,採取されたヒト皮膚をディスパーゼ処理,トリプシン処理,及びコラゲナーゼ処理の順で処理して得られた細胞である,方法。
    The method according to claim 1, wherein
    The human skin-derived stem cell is a cell obtained by treating collected human skin in the order of dispase treatment, trypsin treatment, and collagenase treatment.
  8.  請求項1に記載の方法を用いて培養細胞を製造する工程と,
     前記培養細胞,前記培養工程において得られた培地,及び前記培養工程において得られた分泌物のいずれか1種又は2種以上を含む脊髄損傷疾患の治療剤の製造方法。

     
     
    Producing a cultured cell using the method according to claim 1;
    A method for producing a therapeutic agent for spinal cord injury, comprising any one or two or more of the cultured cells, the culture medium obtained in the culture step, and the secretion obtained in the culture step.


PCT/JP2018/028998 2017-11-10 2018-08-02 Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease WO2019092939A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2019551884A JP7207742B2 (en) 2017-11-10 2018-08-02 Manufacturing method of cultured cells, manufacturing method of therapeutic agent for spinal cord injury disease
US16/763,023 US20210363483A1 (en) 2017-11-10 2018-08-02 Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease
EP18876286.8A EP3708662A4 (en) 2017-11-10 2018-08-02 Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease
AU2018364852A AU2018364852B2 (en) 2017-11-10 2018-08-02 Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease
RU2019110931A RU2730864C1 (en) 2017-11-10 2018-08-02 Method of producing cultured cells and medicinal preparation for treating spinal injuries
CN201880082876.1A CN111566204B (en) 2017-11-10 2018-08-02 Method for producing cultured cells and method for producing therapeutic agent for spinal cord injury disease
CA3080691A CA3080691C (en) 2017-11-10 2018-08-02 Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2017-216849 2017-11-10
JP2017216849 2017-11-10

Publications (1)

Publication Number Publication Date
WO2019092939A1 true WO2019092939A1 (en) 2019-05-16

Family

ID=66439138

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2018/028998 WO2019092939A1 (en) 2017-11-10 2018-08-02 Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease

Country Status (8)

Country Link
US (1) US20210363483A1 (en)
EP (1) EP3708662A4 (en)
JP (1) JP7207742B2 (en)
CN (1) CN111566204B (en)
AU (1) AU2018364852B2 (en)
CA (1) CA3080691C (en)
RU (1) RU2730864C1 (en)
WO (1) WO2019092939A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112795535B (en) * 2021-01-14 2022-06-28 北京臻膳美科技有限公司 Composition for inducing mesenchymal stem cells to release exosomes with specific functions for promoting differentiation of skin epithelial cells and application of composition
CN113599516B (en) * 2021-08-16 2022-02-18 上海蒙彼利生物技术有限公司 Method for preparing exosome and application of pharmaceutical composition thereof in tissue repair

Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS49587B1 (en) 1970-03-10 1974-01-08
JPS549359B2 (en) 1972-03-01 1979-04-24
JPS5620821B2 (en) 1977-08-25 1981-05-15
JPS5856029B2 (en) 1979-12-11 1983-12-13 新日本製鐵株式会社 strip cooling system
JPS6041270B2 (en) 1980-07-29 1985-09-14 日本鋼管株式会社 Method for preventing uneven settlement of piping systems
JPS6137626B2 (en) 1976-03-12 1986-08-25 Ricoh Kk
JPS6210881B2 (en) 1981-08-12 1987-03-09 Daifuku Kk
JP2002531128A (en) * 1998-12-09 2002-09-24 アムジエン・インコーポレーテツド Neurotrophic factor GRNF4
WO2002076976A2 (en) 2001-03-23 2002-10-03 Bayer Corporation Rho-kinase inhibitors
US20030087919A1 (en) 2001-03-23 2003-05-08 Bayer Corporation Rho-kinase inhibitors
WO2003059913A1 (en) 2002-01-10 2003-07-24 Bayer Healthcare Ag Roh-kinase inhibitors
WO2003062227A1 (en) 2002-01-23 2003-07-31 Bayer Pharmaceuticals Corporation Rho-kinase inhibitors
WO2003062225A1 (en) 2002-01-23 2003-07-31 Bayer Pharmaceuticals Corporation Pyrimidine derivatives as rho-kinase inhibitors
WO2004039796A1 (en) 2002-10-28 2004-05-13 Bayer Healthcare Ag Heteroaryloxy-substituted phenylaminopyrimidines as rho-kinase inhibitors
JP2005502377A (en) * 2001-09-20 2005-01-27 アンチキャンサー インコーポレーテッド Hair follicle stem cells expressing nestin
JP2007520207A (en) * 2003-09-03 2007-07-26 リライアンス ライフ サンエンシーズ ピーヴィーティー. リミテッド In vitro generation of GABAergic neurons derived from embryonic stem cells and their use in the treatment of neurological diseases
WO2009148170A1 (en) * 2008-06-06 2009-12-10 独立行政法人理化学研究所 Method for culture of stem cell
JP4385076B2 (en) 2006-01-13 2009-12-16 独立行政法人科学技術振興機構 Media additives, kits and use thereof for serum-free culture of animal cells
WO2011007900A1 (en) * 2009-07-15 2011-01-20 Dezawa Mari Pluripotent stem cell that can be isolated from body tissue
JP2012029684A (en) 2010-06-30 2012-02-16 Cell Aid Kenkyusho:Kk Method for producing cell
JP2012157263A (en) 2011-01-31 2012-08-23 Seems Inc Mesenchymal stem cell originated from human adipose tissue and aimed at treatment of alzheimer's disease
JP2013502915A (en) * 2009-08-25 2013-01-31 セルビシオ アンダルス デ サル Manufacture of artificial tissue by tissue engineering using fibrin and agarose biomaterials
WO2014084085A1 (en) 2012-11-28 2014-06-05 国立大学法人名古屋大学 Prophylactic/therapeutic agent for hearing disorder or cerebellar ataxia
JP6185907B2 (en) 2011-03-30 2017-08-23 セルラー ダイナミクス インターナショナル, インコーポレイテッド Prestimulation of pluripotent stem cells for neural differentiation
JP6191694B2 (en) 2013-08-01 2017-09-06 株式会社ツーセル Cartilage injury therapeutic agent and method for producing the same
JP2017163898A (en) * 2016-03-16 2017-09-21 株式会社日本触媒 Method of culturing neural stem cell, and method of forming neurospheroid

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070269412A1 (en) * 2003-12-02 2007-11-22 Celavie Biosciences, Llc Pluripotent cells
KR100825864B1 (en) * 2006-09-21 2008-04-28 부산대학교 산학협력단 Therapeutic Cell Medicine Comprising Skin Tissue Derived Stem Cell
RU2428475C2 (en) * 2008-12-30 2011-09-10 Андрей Степанович БРЮХОВЕЦКИЙ Stem cell preparation with reprogrammed cell signalling, method for making this preparation and application thereof
CN104487568B (en) * 2012-07-11 2017-08-15 爱姆斯坦生物技术公司 The mesenchymal stem cells of derived from human embryonic stem, method and its application
CN107208045B (en) * 2014-11-25 2018-11-02 宾州研究基金会 Neuroglia cell of human is converted to the chemistry reprogramming for brain and the nerve cell of spinal cord reparation
LU92771B1 (en) * 2015-07-10 2017-01-30 Univ Luxembourg Long-term self-renewing neural stem cells
US20170296587A1 (en) * 2016-04-19 2017-10-19 Creative Medical Health, Inc Stem cell therapy of neurological manifestations of a viral infection

Patent Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS49587B1 (en) 1970-03-10 1974-01-08
JPS549359B2 (en) 1972-03-01 1979-04-24
JPS6137626B2 (en) 1976-03-12 1986-08-25 Ricoh Kk
JPS5620821B2 (en) 1977-08-25 1981-05-15
JPS5856029B2 (en) 1979-12-11 1983-12-13 新日本製鐵株式会社 strip cooling system
JPS6041270B2 (en) 1980-07-29 1985-09-14 日本鋼管株式会社 Method for preventing uneven settlement of piping systems
JPS6210881B2 (en) 1981-08-12 1987-03-09 Daifuku Kk
JP2002531128A (en) * 1998-12-09 2002-09-24 アムジエン・インコーポレーテツド Neurotrophic factor GRNF4
WO2002076976A2 (en) 2001-03-23 2002-10-03 Bayer Corporation Rho-kinase inhibitors
US20030087919A1 (en) 2001-03-23 2003-05-08 Bayer Corporation Rho-kinase inhibitors
US20030125344A1 (en) 2001-03-23 2003-07-03 Bayer Corporation Rho-kinase inhibitors
JP2005502377A (en) * 2001-09-20 2005-01-27 アンチキャンサー インコーポレーテッド Hair follicle stem cells expressing nestin
US20040014755A1 (en) 2002-01-10 2004-01-22 Dhanapalan Nagarathnam Rho-kinase inhibitors
WO2003059913A1 (en) 2002-01-10 2003-07-24 Bayer Healthcare Ag Roh-kinase inhibitors
WO2003062225A1 (en) 2002-01-23 2003-07-31 Bayer Pharmaceuticals Corporation Pyrimidine derivatives as rho-kinase inhibitors
US20040002507A1 (en) 2002-01-23 2004-01-01 Bayer Corporation Rho-kinase inhibitors
US20040002508A1 (en) 2002-01-23 2004-01-01 Bayer Corporation Rho-kinase inhibitors
WO2003062227A1 (en) 2002-01-23 2003-07-31 Bayer Pharmaceuticals Corporation Rho-kinase inhibitors
US20050192304A1 (en) 2002-01-23 2005-09-01 Dhanapalan Nagarathnam Rho-kinase inhibitors
US20050209261A1 (en) 2002-01-23 2005-09-22 Dhanapalan Nagarathnam Rho-kinase inhibitors
WO2004039796A1 (en) 2002-10-28 2004-05-13 Bayer Healthcare Ag Heteroaryloxy-substituted phenylaminopyrimidines as rho-kinase inhibitors
JP2007520207A (en) * 2003-09-03 2007-07-26 リライアンス ライフ サンエンシーズ ピーヴィーティー. リミテッド In vitro generation of GABAergic neurons derived from embryonic stem cells and their use in the treatment of neurological diseases
JP4385076B2 (en) 2006-01-13 2009-12-16 独立行政法人科学技術振興機構 Media additives, kits and use thereof for serum-free culture of animal cells
WO2009148170A1 (en) * 2008-06-06 2009-12-10 独立行政法人理化学研究所 Method for culture of stem cell
WO2011007900A1 (en) * 2009-07-15 2011-01-20 Dezawa Mari Pluripotent stem cell that can be isolated from body tissue
JP2013502915A (en) * 2009-08-25 2013-01-31 セルビシオ アンダルス デ サル Manufacture of artificial tissue by tissue engineering using fibrin and agarose biomaterials
JP2012029684A (en) 2010-06-30 2012-02-16 Cell Aid Kenkyusho:Kk Method for producing cell
JP2012157263A (en) 2011-01-31 2012-08-23 Seems Inc Mesenchymal stem cell originated from human adipose tissue and aimed at treatment of alzheimer's disease
JP6185907B2 (en) 2011-03-30 2017-08-23 セルラー ダイナミクス インターナショナル, インコーポレイテッド Prestimulation of pluripotent stem cells for neural differentiation
WO2014084085A1 (en) 2012-11-28 2014-06-05 国立大学法人名古屋大学 Prophylactic/therapeutic agent for hearing disorder or cerebellar ataxia
JP6191694B2 (en) 2013-08-01 2017-09-06 株式会社ツーセル Cartilage injury therapeutic agent and method for producing the same
JP2017163898A (en) * 2016-03-16 2017-09-21 株式会社日本触媒 Method of culturing neural stem cell, and method of forming neurospheroid

Non-Patent Citations (35)

* Cited by examiner, † Cited by third party
Title
ANSORENA E ET AL.: "Injectable alginate hydrogel loaded with GDNF promotes functional recovery in a hemisection model of spinal cord injury", INT J PHARM., vol. 455, no. 1-2, 2013, pages 148 - 58, XP028699749, DOI: 10.1016/j.ijpharm.2013.07.045
BIEMASKIE J ET AL.: "Skin-derived precursors generate myelinating Schwann cells that promote remyelination and functional recovery after contusion spinal cord injury", J NEUROSCIENCE, vol. 27, no. 36, 2007, pages 9545 - 9559, XP055331841, DOI: 10.1523/JNEUROSCI.1930-07.2007
CAO L ET AL.: "Olfactory ensheathing cells genetically modified to secrete GDNF to promote spinal cord repair", BRAIN, vol. 127, March 2004 (2004-03-01), pages 535 - 49
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 364590-63-6
CHEN JKTAIPALE JYOUNG KEMAITI TBEACHY PA, PROC NATL ACAD SCI USA., vol. 99, no. 22, 29 October 2002 (2002-10-29), pages 14071 - 6
CHEN, J, K. ET AL.: "Small molecule modulation of Smoothened activity", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 99, no. 22, 29 October 2002 (2002-10-29), pages 14071 - 14076, XP002251705 *
CHEN, W. ET AL.: "Activity-dependent internalization of smoothened mediated by beta-arrestin 2 and GRK2.", SCIENCE, vol. 306, no. 5705, 24 December 2004 (2004-12-24), pages 2257 - 2260, XP055607826 *
CHEN, W.REN, X. R.NELSON, C. D.BARAK, L. S.CHEN, J. K.BEACHY, P. A.DE SAUVAGE, F.LEFKOWITZ, R. J., SCIENCE, vol. 306, no. 5705, 2004, pages 2257 - 2260
CHOU AK ET AL.: "Adenoviral-mediated glial cell line-derived neurotrophic factor gene transfer has a protective effect on sciatic nerve following constriction-induced spinal cord injury", PLOS ONE, vol. 9, no. 3, 2014, pages e92264
CODEGA P ET AL.: "Prospective identification and purification of quiescent adult neural stem cells from their in vivo niche", NEURON, vol. 82, no. 3, 2014, pages 545 - 59, XP028654862, DOI: 10.1016/j.neuron.2014.02.039
DECAROLIS NA ET AL.: "In vivo contribution of nestin-and GLAST-lineage cells to adult hippocampal neurogenesis", HIPPOCAMPUS, vol. 23, no. 8, 2013, pages 708 - 719
DENG LX ET AL.: "A novel growth-promoting pathway formed by GDNF-overexpressing Schwann cells promotes propriospinal axonal regeneration, synapse formation, and partial recovery of function after spinal cord injury", J NEUROSCI, vol. 33, no. 13, 27 March 2013 (2013-03-27), pages 5655 - 67
DETLOFF MR ET AL.: "Acute exercise prevents the development of neuropathic pain and the sprouting of non-peptidergic (GDNF- and artemin-responsive) c-fibers after spinal cord injury", EXP NEUROL, vol. 255, 2014, pages 38 - 48
DOLBEARE D ET AL.: "Restriction of axonal retraction and promotion of axonal regeneration by chronically injured neurons after intraspinal treatment with glial cell line-derived neurotrophic factor (GDNF", J NEUROTRAUMA, vol. 20, no. 11, 2003, pages 1251 - 61
GUZEN FP ET AL.: "Glial cell line-derived neurotrophic factor added to a sciatic nerve fragment grafted in a spinal cord gap ameliorates motor impairments in rats and increases local axonal growth", RESTOR NEUROL NEUROSCI, vol. 27, no. 1, 2009, pages 1 - 16
INOUE H ET AL.: "Improvement of hind-limb paralysis following traumatic spinal cord injury in rats by grafting normal human keratinocytes: new cell-therapy strategy for nerve regeneration", J ARTIF ORGANS, vol. 14, no. 4, 2011, pages 375 - 80, XP019987750, DOI: 10.1007/s10047-011-0598-0
INOUE H ET AL.: "Improvement of hind-paralysis following traumatic spinal cord injury in rats by grafting normal human keratinocytes: new cell-therapy strategy for nerve regeneration", J ARTIF ORGANS, vol. 14, no. 4, 2011, pages 375 - 80, XP019987750, DOI: 10.1007/s10047-011-0598-0
ISHIZAKI ET AL., MOL. PHARMACOL., vol. 57, 2000, pages 976 - 983
KAO CH ET AL.: "Exogenous administration of glial cell line-derived neurotrophic factor improves recovery after spinal cord injury", RESUSCITATION, vol. 77, no. 3, 2008, pages 395 - 400, XP022667026, DOI: 10.1016/j.resuscitation.2008.01.023
KIM MS ET AL.: "Perspectives on Tissue-Engineered Nerve Regeneration for the Treatment of Spinal Cord Injury", TISSUE ENG PART, vol. 20, no. 13-14, 2014, pages 1781 - 3
KOELSCH A ET AL.: "Transgene-mediated GDNF expression enhances synaptic connectivity and GAB A transmission to improve functional outcome after spinal cord contusion", J NEUROCHEM, vol. 113, no. 1, 2010, pages 143 - 52
LU Y ET AL.: "Glial cell line-derived neurotrophic factor-transfected placenta-derived versus bone marrow-derived mesenchymal cells for treating spinal cord injury", MED SCI MONIT, vol. 23, 2017, pages 1800 - 1811
NAKAJIMA ET AL., CANCER CHEMOTHER. PHARMACOL., vol. 52, no. 4, 2003, pages 319 - 324
NARUMIYA ET AL., METHODS ENZYMOL., vol. 325, 2000, pages 273 - 284
SASAKI ET AL., PHARMACOL. THER., vol. 93, 2002, pages 225 - 232
See also references of EP3708662A4
SINHA SCHEN JK, NAT CHEM BIOL., vol. 2, no. 1, January 2006 (2006-01-01), pages 29 - 30
SOWA NA ET AL.: "Ecto-5'-nucleotidase (CD73) inhibits nociception by hydrolyzing AMP to adenosine in nociceptive circuits", J NEUROSCI, vol. 30, no. 6, 10 February 2010 (2010-02-10), pages 2235 - 2244
STEPHEN J. A. DAVIEES ET AL.: "Transplantaion of specific human astrocytes promotes functional recovery after spinal cord injury", PLOS ONE, vol. 6, no. 3, 2011, pages e17328
SUFAN WU ET AL.: "Bone marrow stromal cells enhance differentiation of cocultured neurosphere cells and promote regeneration of injured spinal cord", J NEUROSCI RES, vol. 72, no. 3, 2003, pages 343 - 51
TANG XQ ET AL.: "Adenovirus-mediated delivery of GDNF ameliorates corticospinal neuronal atrophy and motor function deficits in rats with spinal cord injury", NEUROREPORT, vol. 15, no. 3, 2004, pages 425 - 9
TSUJI O ET AL.: "Therapeutic potential of appropriately evaluated self-induced pluripotent stem cells for spinal cord injury", PNAS, vol. 107, no. 28, 2010, pages 12704 - 12709
UENATA ET AL., NATURE, vol. 389, 1997, pages 990 - 994
ZHAO YZ ET AL.: "Thermosensitive heparin-poloxamer hydrogels enhance the effects of GDNF on neuronal circuit remodeling and neuroprotection after spinal cord injury", J BIOMED MATER RES A, vol. 5, no. 10, 2017, pages 2816 - 2829
ZHOU HL ET AL.: "Changes in Glial cell line-derived neurotrophic factor expression in the rostral and caudal stumps of the transected adult rat spinal cord", NEUROCHEM RES, vol. 33, no. 5, 2008, pages 927 - 37, XP019579149

Also Published As

Publication number Publication date
RU2730864C1 (en) 2020-08-26
AU2018364852A1 (en) 2020-05-14
JPWO2019092939A1 (en) 2021-01-21
CN111566204A (en) 2020-08-21
CA3080691C (en) 2022-11-15
EP3708662A4 (en) 2021-08-25
AU2018364852B2 (en) 2022-06-30
EP3708662A1 (en) 2020-09-16
CN111566204B (en) 2024-03-26
CA3080691A1 (en) 2019-05-16
JP7207742B2 (en) 2023-01-18
US20210363483A1 (en) 2021-11-25

Similar Documents

Publication Publication Date Title
US20220016110A1 (en) Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
EP2099901B1 (en) Use of a composition contaning human umbilical cord blood-derived mesenchymal stem cell for inducing differentiation and proliferation of neural precursor cells or neural stem cells to neural cells
EP2331679B1 (en) Ischemic tissue cell therapy
KR20210107144A (en) Treatment of spinal cord injury and traumatic brain injury using placental stem cells
JPWO2006041088A1 (en) Brain transitional bone marrow progenitor cells
WO2014065348A1 (en) Novel method for treating spinal cord injury using hmgb1 fragment
JP7045670B2 (en) Reduction and treatment of ischemia-reperfusion lung injury with pluripotent stem cells
WO2019092939A1 (en) Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease
WO2017110425A1 (en) Therapeutic agent for liver disease and method for treating liver disease
Zhang et al. Exosomes derived from bone marrow mesenchymal stem cells pretreated with decellularized extracellular matrix enhance the alleviation of osteoarthritis through miR‐3473b/phosphatase and tensin homolog axis
JP7506905B2 (en) Additive
WO2024063020A1 (en) Intervertebral disc therapeutic agent
US20200108100A1 (en) Neural stem cell therapy for stroke
KR20110118084A (en) Multipotent adult stem cells derived from tympanic membrane tissue, preparation method thereof and differentiated cells from the stem cells

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18876286

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3080691

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2019551884

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018364852

Country of ref document: AU

Date of ref document: 20180802

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2018876286

Country of ref document: EP

Effective date: 20200610